Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Clin Invest ; 134(10)2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38512451

RESUMO

Lactylation has been recently identified as a new type of posttranslational modification occurring widely on lysine residues of both histone and nonhistone proteins. The acetyltransferase p300 is thought to mediate protein lactylation, yet the cellular concentration of the proposed lactyl-donor, lactyl-coenzyme A, is about 1,000 times lower than that of acetyl-CoA, raising the question of whether p300 is a genuine lactyltransferase. Here, we report that alanyl-tRNA synthetase 1 (AARS1) moonlights as a bona fide lactyltransferase that directly uses lactate and ATP to catalyze protein lactylation. Among the candidate substrates, we focused on the Hippo pathway, which has a well-established role in tumorigenesis. Specifically, AARS1 was found to sense intracellular lactate and translocate into the nucleus to lactylate and activate the YAP-TEAD complex; and AARS1 itself was identified as a Hippo target gene that forms a positive-feedback loop with YAP-TEAD to promote gastric cancer (GC) cell proliferation. Consistently, the expression of AARS1 was found to be upregulated in GC, and elevated AARS1 expression was found to be associated with poor prognosis for patients with GC. Collectively, this work found AARS1 with lactyltransferase activity in vitro and in vivo and revealed how the metabolite lactate is translated into a signal of cell proliferation.


Assuntos
Transdução de Sinais , Neoplasias Gástricas , Fatores de Transcrição , Proteínas de Sinalização YAP , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/enzimologia , Humanos , Animais , Proteínas de Sinalização YAP/metabolismo , Proteínas de Sinalização YAP/genética , Linhagem Celular Tumoral , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/genética , Ácido Láctico/metabolismo , Aminoacil-tRNA Sintetases/metabolismo , Aminoacil-tRNA Sintetases/genética , Regulação Neoplásica da Expressão Gênica , Proliferação de Células
3.
Adv Sci (Weinh) ; 8(17): e2004850, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34240584

RESUMO

Elevated Wnt/ß-catenin signaling has been commonly associated with tumorigenesis especially colorectal cancer (CRC). Here, an MST4-pß-cateninThr40 signaling axis essential for intestinal stem cell (ISC) homeostasis and CRC development is uncovered. In response to Wnt3a stimulation, the kinase MST4 directly phosphorylates ß-catenin at Thr40 to block its Ser33 phosphorylation by GSK3ß. Thus, MST4 mediates an active process that prevents ß-catenin from binding to and being degraded by ß-TrCP, leading to accumulation and full activation of ß-catenin. Depletion of MST4 causes loss of ISCs and inhibits CRC growth. Mice bearing either MST4T178E mutation with constitutive kinase activity or ß-cateninT40D mutation mimicking MST4-mediated phosphorylation show overly increased ISCs/CSCs and exacerbates CRC. Furthermore, the MST4-pß-cateninThr40 axis is upregulated and correlated with poor prognosis of human CRC. Collectively, this work establishes a previously undefined machinery for ß-catenin activation, and further reveals its function in stem cell and tumor biology, opening new opportunities for targeted therapy of CRC.


Assuntos
Carcinogênese/genética , Neoplasias Colorretais/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/metabolismo , Animais , Neoplasias Colorretais/genética , Modelos Animais de Doenças , Feminino , Humanos , Intestinos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/genética , Células-Tronco/metabolismo , beta Catenina/genética
4.
Genome Med ; 13(1): 58, 2021 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-33853662

RESUMO

BACKGROUND: Aberrant changes in epigenetic mechanisms such as histone modifications play an important role in cancer progression. PRMT1 which triggers asymmetric dimethylation of histone H4 on arginine 3 (H4R3me2a) is upregulated in human colorectal cancer (CRC) and is essential for cell proliferation. However, how this dysregulated modification might contribute to malignant transitions of CRC remains poorly understood. METHODS: In this study, we integrated biochemical assays including protein interaction studies and chromatin immunoprecipitation (ChIP), cellular analysis including cell viability, proliferation, colony formation, and migration assays, clinical sample analysis, microarray experiments, and ChIP-Seq data to investigate the potential genomic recognition pattern of H4R3me2s in CRC cells and its effect on CRC progression. RESULTS: We show that PRMT1 and SMARCA4, an ATPase subunit of the SWI/SNF chromatin remodeling complex, act cooperatively to promote colorectal cancer (CRC) progression. We find that SMARCA4 is a novel effector molecule of PRMT1-mediated H4R3me2a. Mechanistically, we show that H4R3me2a directly recruited SMARCA4 to promote the proliferative, colony-formative, and migratory abilities of CRC cells by enhancing EGFR signaling. We found that EGFR and TNS4 were major direct downstream transcriptional targets of PRMT1 and SMARCA4 in colon cells, and acted in a PRMT1 methyltransferase activity-dependent manner to promote CRC cell proliferation. In vivo, knockdown or inhibition of PRMT1 profoundly attenuated the growth of CRC cells in the C57BL/6 J-ApcMin/+ CRC mice model. Importantly, elevated expression of PRMT1 or SMARCA4 in CRC patients were positively correlated with expression of EGFR and TNS4, and CRC patients had shorter overall survival. These findings reveal a critical interplay between epigenetic and transcriptional control during CRC progression, suggesting that SMARCA4 is a novel key epigenetic modulator of CRC. Our findings thus highlight PRMT1/SMARCA4 inhibition as a potential therapeutic intervention strategy for CRC. CONCLUSION: PRMT1-mediated H4R3me2a recruits SMARCA4, which promotes colorectal cancer progression by enhancing EGFR signaling.


Assuntos
Arginina/metabolismo , Neoplasias Colorretais/metabolismo , DNA Helicases/metabolismo , Progressão da Doença , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Sulfato de Dextrana , Receptores ErbB/metabolismo , Humanos , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Prognóstico , Tensinas/metabolismo , Transcrição Gênica , Regulação para Cima
5.
Cancer Res ; 81(11): 3036-3050, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33619115

RESUMO

Most primary liver cancer (PLC) cases progress mainly due to underlying chronic liver inflammation, yet the underlying mechanisms of inflammation-mediated PLC remain unclear. Here we uncover a TNF receptor II (TNFR2)-hnRNPK-YAP signaling axis in hepatic progenitor cells (HPC) essential for PLC development. TNFR2, but not TNF receptor I (TNFR1), was required for TNFα-induced activation of YAP during malignant transformation of HPCs and liver tumorigenesis. Mechanistically, heterogeneous nuclear ribonuclear protein K (hnRNPK) acted downstream of TNFα-TNFR2 signaling to directly interact with and stabilize YAP on target gene promoters genome-wide, therefore coregulating the expression of YAP target genes. Single-cell RNA sequencing confirmed the association of TNFR2-hnRNPK with YAP expression and the pathologic importance of HPC. Accordingly, expressions of TNFR2, hnRNPK, and YAP were all upregulated in PLC tissues and were strongly associated with poor prognosis of PLC including patient survival. Collectively, this study clarifies the differential roles of TNFRs in HPC-mediated tumorigenesis, uncovering a TNFR2-hnRNPK-centered mechanistic link between the TNFα-mediated inflammatory milieu and YAP activation in HPCs during PLC development. SIGNIFICANCE: This work defines how hnRNPK links TNFα signaling and Hippo pathway transcription coactivator YAP in hepatic progenitor cells during primary liver tumorigenesis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Células-Tronco/patologia , Fatores de Transcrição/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proteínas de Ciclo Celular/genética , Proliferação de Células , Feminino , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/genética , Humanos , Neoplasias Hepáticas/genética , Prognóstico , Ratos , Ratos Sprague-Dawley , Receptores Tipo II do Fator de Necrose Tumoral/genética , Transdução de Sinais , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
6.
Theranostics ; 10(10): 4437-4452, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32292506

RESUMO

The proto-oncogene c-Myc regulates multiple biological processes mainly through selectively activating gene expression. However, the mechanisms underlying c-Myc-mediated gene repression in the context of cancer remain less clear. This study aimed to clarify the role of PRMT5 in the transcriptional repression of c-Myc target genes in gastric cancer. Methods: Immunohistochemistry was used to evaluate the expression of PRMT5, c-Myc and target genes in gastric cancer patients. PRMT5 and c-Myc interaction was assessed by immunofluorescence, co-immunoprecipitation and GST pull-down assays. Bioinformatics analysis, immunoblotting, real-time PCR, chromatin immunoprecipitation, and rescue experiments were used to evaluate the mechanism. Results: We found that c-Myc directly interacts with protein arginine methyltransferase 5 (PRMT5) to transcriptionally repress the expression of a cohort of genes, including PTEN, CDKN2C (p18INK4C), CDKN1A (p21CIP1/WAF1), CDKN1C (p57KIP2) and p63, to promote gastric cancer cell growth. Specifically, we found that PRMT5 was required to promote gastric cancer cell growth in vitro and in vivo, and for transcriptional repression of this cohort of genes, which was dependent on its methyltransferase activity. Consistently, the promoters of this gene cohort were enriched for both PRMT5-mediated symmetric di-methylation of histone H4 on Arg 3 (H4R3me2s) and c-Myc, and c-Myc depletion also upregulated their expression. H4R3me2s also colocalized with the c-Myc-binding E-box motif (CANNTG) on these genes. We show that PRMT5 directly binds to c-Myc, and this binding is required for transcriptional repression of the target genes. Both c-Myc and PRMT5 expression levels were upregulated in primary human gastric cancer tissues, and their expression levels inversely correlated with clinical outcomes. Conclusions: Taken together, our study reveals a novel mechanism by which PRMT5-dependent transcriptional repression of c-Myc target genes is required for gastric cancer progression, and provides a potential new strategy for therapeutic targeting of gastric cancer.


Assuntos
Adenocarcinoma/metabolismo , Histonas/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Neoplasias Gástricas/metabolismo , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Progressão da Doença , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Metilação , Regiões Promotoras Genéticas , Proto-Oncogene Mas , Neoplasias Gástricas/patologia
7.
Aging (Albany NY) ; 12(2): 1304-1321, 2020 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-31982864

RESUMO

Aurora kinase B (AURKB) triggers the phosphorylation of serine 10 on histone H3 (H3S10ph), which is important for chromosome condensation and cytokinesis during mitosis in mammals. However, how exactly AURKB controls cell cycle and contributes to tumorigenesis as an oncoprotein under pathological conditions remains largely unknown. Here, we report that AURKB promotes gastric cancer cell proliferation in vitro and in vivo. Silencing AURKB expression inhibits gastric cell proliferation and arrests the cell cycle in G2/M phase. We demonstrate that cyclin D1 (CCND1) is a direct downstream target of AURKB that plays a key role in gastric cancer cell proliferation. AURKB is able to activate the expression of CCND1 through mediating H3S10ph in the promoter of the CCND1 gene. Furthermore, we show that AZD1152, a specific inhibitor of AURKB, can suppress the expression of CCND1 in the gastric cancer cells and inhibit cell proliferation in vitro and in vivo. Importantly, we found that high AURKB and CCND1 expression levels are correlated with shorter overall survival of gastric cancer patients. This study demonstrates that AURKB promotes gastric tumorigenesis potentially through epigenetically activating CCND1 expression, suggesting AURKB as a promising therapeutic target in gastric cancer.


Assuntos
Aurora Quinase B/metabolismo , Ciclina D1/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Aurora Quinase B/genética , Biomarcadores Tumorais , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1/metabolismo , Ativação Enzimática , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Fosforilação , Prognóstico , Ligação Proteica , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/patologia
8.
FEBS Open Bio ; 9(12): 2159-2169, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31705785

RESUMO

Platelets are implicated in the pathophysiology of breast and other cancers through their role in exchanging biomolecules with tumor cells in the tumor microenvironment. Such exchange results in tumor-educated platelets with altered RNA expression profiles. Multiple lines of evidence indicate that platelet RNA profiles may be suitable as diagnostic biomarkers for cancer-related biological processes. In this study, we characterized the gene expression signatures of platelets in breast cancer (BC) by high-throughput sequencing and quantitative real-time RT-PCR. Our results indicate that the expression of TPM3 (tropomyosin 3) mRNA is significantly elevated in platelets from patients with BC compared with age-matched healthy control subjects. Furthermore, up-regulation of TPM3 mRNA in platelets was found to be significantly correlated with metastasis in patients with BC. Finally, we report that platelet TPM3 mRNA is delivered into BC cells through microvesicles and leads to enhanced migrative phenotype of BC cells. In summary, our findings suggest that the transfer of platelet TPM3 mRNA into cancer cells via microvesicles promotes cancer cell migration, and thus platelet-derived TPM3 mRNA may be a suitable biomarker for early diagnosis of metastatic BC.


Assuntos
Neoplasias da Mama/genética , Micropartículas Derivadas de Células/genética , Tropomiosina/metabolismo , Adulto , Idoso , Biomarcadores Tumorais , Plaquetas/fisiologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/fisiopatologia , Linhagem Celular Tumoral , Movimento Celular , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Pessoa de Meia-Idade , Metástase Neoplásica/genética , Metástase Neoplásica/fisiopatologia , Processos Neoplásicos , RNA Mensageiro/genética , Transcriptoma/genética , Tropomiosina/fisiologia , Microambiente Tumoral/fisiologia
9.
J Biol Chem ; 293(45): 17454-17463, 2018 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-30257864

RESUMO

Protein arginine methyltransferase 5 (PRMT5) is a member of the arginine methyltransferase protein family that critically mediates the symmetric dimethylation of Arg-3 at histone H4 (H4R3me2s) and is involved in many key cellular processes, including hematopoiesis. However, the post-translational modifications (PTMs) of PRMT5 that may affect its biological functions remain less well-understood. In this study, using MS analyses, we found that PRMT5 itself is methylated in human erythroleukemia Lys-562 cells. Biochemical assays revealed that coactivator-associated arginine methyltransferase 1 (CARM1) interacts directly with and methylates PRMT5 at Arg-505 both in vivo and in vitro. Substitutions at Arg-505 significantly reduced PRMT5's methyltransferase activity, decreased H4R3me2s enrichment at the γ-globin gene promoter, and increased the expression of the γ-globin gene in Lys-562 cells. Moreover, CARM1 knockdown consistently reduced PRMT5 activity and activated γ-globin gene expression. Importantly, we show that CARM1-mediated methylation of PRMT5 is essential for the intracellular homodimerization of PRMT5 to its active form. These results thus reveal a critical PTM of PRMT5 that represses human γ-globin gene expression. We conclude that CARM1-mediated asymmetric methylation of PRMT5 is critical for its dimerization and methyltransferase activity leading to the repression of γ-globin expression. Given PRMT5's crucial role in diverse cellular processes, these findings may inform strategies for manipulating its methyltransferase activity for managing hemoglobinopathy or cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Leucemia Eritroblástica Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , gama-Globinas/biossíntese , Linhagem Celular Tumoral , Metilação de DNA/genética , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patologia , Proteínas de Neoplasias/genética , Proteína-Arginina N-Metiltransferases/genética , gama-Globinas/genética
10.
Oncol Lett ; 15(5): 6123-6130, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29616094

RESUMO

Methylation of histone H4 lysine 20 (H4K20) has been associated with cancer. However, the functions of the histone methyltransferases that trigger histone H4K20 methylation in cancers, including suppressor of variegation 4-20 homolog 1 (Suv4-20h1), remain elusive. In the present study, it was demonstrated that the knockdown of the histone H4K20 methyltransferase Suv4-20h1 resulted in growth inhibition in chronic myeloid leukemia K562 cells. Disruption of Suv4-20h1 expression induced G1 arrest in the cell cycle and increased expression levels of cyclin dependent kinase inhibitor 1A (p21WAF1/CIP1), an essential cell cycle protein involved in checkpoint regulation. Chromatin immunoprecipitation analysis demonstrated that Suv4-20h1 directly binds to the promoter of the p21 gene and that methylation of histone H4K20 correlates with repression of p21 expression. Thus, these data suggest that Suv4-20h1 is important for the regulation of the cell cycle in K562 cells and may be a potential therapeutic target for leukemia.

11.
Nat Commun ; 8(1): 928, 2017 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-29030587

RESUMO

N-α-acetyltransferase D (NatD) mediates N-α-terminal acetylation (Nt-acetylation) of histone H4 known to be involved in cell growth. Here we report that NatD promotes the migratory and invasive capabilities of lung cancer cells in vitro and in vivo. Depletion of NatD suppresses the epithelial-to-mesenchymal transition (EMT) of lung cancer cells by directly repressing the expression of transcription factor Slug, a key regulator of EMT. We found that Nt-acetylation of histone H4 antagonizes histone H4 serine 1 phosphorylation (H4S1ph), and that downregulation of Nt-acetylation of histone H4 facilitates CK2α binding to histone H4 in lung cancer cells, resulting in increased H4S1ph and epigenetic reprogramming to suppress Slug transcription to inhibit EMT. Importantly, NatD is commonly upregulated in primary human lung cancer tissues where its expression level correlates with Slug expression, enhanced invasiveness, and poor clinical outcomes. These findings indicate that NatD is a crucial epigenetic modulator of cell invasion during lung cancer progression.NatD is an acetyltransferase responsible for N-α-terminal acetylation of the histone H4 and H2A and has been linked to cell growth. Here the authors show that NatD-mediated acetylation of histone H4 serine 1 competes with the phosphorylation by CK2α at the same residue thus leading to the upregulation of Slug and tumor progression.


Assuntos
Adenocarcinoma/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias Pulmonares/metabolismo , Acetiltransferase N-Terminal D/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Células A549 , Animais , Caseína Quinase II/metabolismo , Movimento Celular , China/epidemiologia , Transição Epitelial-Mesenquimal , Feminino , Células HEK293 , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/mortalidade , Masculino , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica , Fosforilação
12.
Am J Hum Genet ; 101(1): 130-138, 2017 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-28669403

RESUMO

A delayed fetal-to-adult hemoglobin (Hb) switch ameliorates the severity of ß-thalassemia and sickle cell disease. The molecular mechanism underlying the epigenetic dysregulation of the switch is unclear. To explore the potential cis-variants responsible for the Hb switching, we systematically analyzed an 80-kb region spanning the ß-globin cluster using capture-based next-generation sequencing of 1142 Chinese ß-thalassemia persons and identified 31 fetal hemoglobin (HbF)-associated haplotypes of the selected 28 tag regulatory single-nucleotide polymorphisms (rSNPs) in seven linkage disequilibrium (LD) blocks. A Ly1 antibody reactive (LYAR)-binding motif disruptive rSNP rs368698783 (G/A) from LD block 5 in the proximal promoter of hemoglobin subunit gamma 1 (HBG1) was found to be a significant predictor for ß-thalassemia clinical severity by epigenetic-mediated variant-dependent HbF elevation. We found this rSNP accounted for 41.6% of ß-hemoglobinopathy individuals as an ameliorating factor in a total of 2,738 individuals from southern China and Thailand. We uncovered that the minor allele of the rSNP triggers the attenuation of LYAR and two repressive epigenetic regulators DNA methyltransferase 3 alpha (DNMT3A) and protein arginine methyltransferase 5 (PRMT5) from the HBG promoters, mediating allele-biased γ-globin elevation by facilitating demethylation of HBG core promoter CpG sites in erythroid progenitor cells from ß-thalassemia persons. The present study demonstrates that this common rSNP in the proximal Aγ-promoter is a major genetic modifier capable of ameliorating the severity of thalassemia major through the epigenetic-mediated regulation of the delayed fetal-to-adult Hb switch and provides potential targets for the treatment of ß-hemoglobinopathy.


Assuntos
Epigênese Genética , Hemoglobina Fetal/genética , Variação Genética , Talassemia beta/genética , Sequência de Bases , Células Cultivadas , Pré-Escolar , Estudos de Coortes , Feminino , Hemoglobina Fetal/metabolismo , Humanos , Lactente , Células K562 , Masculino , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas , Modelos de Riscos Proporcionais , Transcrição Gênica , Ativação Transcricional/genética
13.
J Biol Chem ; 292(12): 4811-4817, 2017 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-28154185

RESUMO

Production of hemoglobin during development is tightly regulated. For example, expression from the human ß-globin gene locus, comprising ß-, δ-, ϵ-, and γ-globin genes, switches from ϵ-globin to γ-globin during embryonic development and then from γ-globin to ß-globin after birth. Expression of human ϵ-globin in mice has been shown to ameliorate anemia caused by ß-globin mutations, including those causing ß-thalassemia and sickle cell disease, raising the prospect that reactivation of ϵ-globin expression could be used in managing these conditions in humans. Although the human globin genes are known to be regulated by a variety of multiprotein complexes containing enzymes that catalyze epigenetic modifications, the exact mechanisms controlling ϵ-globin gene silencing remain elusive. Here we found that the heterochromatin protein HP1γ, a multifunctional chromatin- and DNA-binding protein with roles in transcriptional activation and elongation, represses ϵ-globin expression by interacting with a histone-modifying enzyme, lysine methyltransferase SUV4-20h2. Silencing of HP1γ expression markedly decreased repressive histone marks and the multimethylation of histone H3 lysine 9 and H4 lysine 20, leading to a significant decrease in DNA methylation at the proximal promoter of the ϵ-globin gene and greatly increased ϵ-globin expression. In addition, using chromatin immunoprecipitation, we showed that SUV4-20h2 facilitates the deposition of HP1γ on the ϵ-globin-proximal promoter. Thus, these data indicate that HP1γ is a novel epigenetic repressor of ϵ-globin gene expression and provide a potential strategy for targeted therapies for ß-thalassemia and sickle cell disease.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Repressão Epigenética , Globinas épsilon/genética , Linhagem Celular , Homólogo 5 da Proteína Cromobox , Metilação de DNA , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Regiões Promotoras Genéticas , Ativação Transcricional
15.
Cancer Res ; 75(21): 4593-604, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26333808

RESUMO

Colorectal cancer pathogenesis remains incompletely understood. Here, we report that the heterochromatin protein HP1γ is upregulated commonly in human colorectal cancer, where it promotes cell proliferation in vitro and in vivo. Gene-expression and promoter-binding experiments demonstrated that HP1γ directly regulated CDKN1A (p21(Waf1/Cip1)) in a manner associated with methylation of histone H3K9 on its promoter. We identified miR-30a as a tumor-suppressive microRNA that targets HP1γ in vitro and in vivo to specifically suppress the growth of colorectal cancer in mouse xenograft models. MiR-30a was widely downregulated in primary human colorectal cancer tissues, where its expression correlated inversely with high levels of HP1γ protein. Our results identify a new miR-30a/HP1γ/p21 regulatory axis controlling colorectal cancer development, which may offer prognostic and therapeutic opportunities.


Assuntos
Transformação Celular Neoplásica/patologia , Proteínas Cromossômicas não Histona/metabolismo , Neoplasias Colorretais/patologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , MicroRNAs/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Proteínas Cromossômicas não Histona/genética , Neoplasias Colorretais/genética , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Metilação de DNA , Progressão da Doença , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HEK293 , Histonas/genética , Histonas/metabolismo , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA Interferente Pequeno , Transplante Heterólogo
16.
Oncotarget ; 6(32): 32890-901, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26413750

RESUMO

Colorectal cancer (CRC) is one of the leading causes of cancer-related death worldwide. However, the molecular mechanisms of CRC pathogenesis are not fully understood. In this study, we report the characterization of LYAR (Ly-1 antibody reactive clone) as a key regulator of the migration and invasion of human CRC cells. Immunohistochemistry analysis demonstrated that LYAR is expressed at a higher level in metastatic CRC tissues. We found that LYAR promoted the migratory and invasive capabilities of CRC cells. Gene expression profile analysis of CRC cells showed that LGALS1, which encodes the galectin-1 protein, was a potential target of LYAR. The ChIP assay and gene reporter assays indicated that LYAR directly bound to the LGALS1 promoter. The ectopic expression of galectin-1 partially restored the mobile potential of LYAR knocked-down cells, which suggests that galectin-1 contributed to the LYAR-promoted cell migration and invasion of CRC cells. Thus, this study revealed a novel mechanism by which the transcription factor LYAR may promote tumor cell migration and invasion by upregulating galectin-1 gene expression in CRC.


Assuntos
Movimento Celular , Neoplasias Colorretais/metabolismo , Proteínas de Ligação a DNA/metabolismo , Galectina 1/metabolismo , Proteínas Nucleares/metabolismo , Sítios de Ligação , Imunoprecipitação da Cromatina , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Proteínas de Ligação a DNA/genética , Galectina 1/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Células HCT116 , Humanos , Imuno-Histoquímica , Invasividade Neoplásica , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Interferência de RNA , Transfecção , Regulação para Cima
17.
Nucleic Acids Res ; 42(15): 9740-52, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25092918

RESUMO

Human globin gene expression during development is modulated by transcription factors in a stage-dependent manner. However, the mechanisms controlling the process are still largely unknown. In this study, we found that a nuclear protein, LYAR (human homologue of mouse Ly-1 antibody reactive clone) directly interacted with the methyltransferase PRMT5 which triggers the histone H4 Arg3 symmetric dimethylation (H4R3me2s) mark. We found that PRMT5 binding on the proximal γ-promoter was LYAR-dependent. The LYAR DNA-binding motif (GGTTAT) was identified by performing CASTing (cyclic amplification and selection of targets) experiments. Results of EMSA and ChIP assays confirmed that LYAR bound to a DNA region corresponding to the 5'-untranslated region of the γ-globin gene. We also found that LYAR repressed human fetal globin gene expression in both K562 cells and primary human adult erythroid progenitor cells. Thus, these data indicate that LYAR acts as a novel transcription factor that binds the γ-globin gene, and is essential for silencing the γ-globin gene.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Nucleares/metabolismo , gama-Globinas/genética , Regiões 5' não Traduzidas , Animais , Sequência de Bases , Diferenciação Celular/genética , Células Cultivadas , Sequência Consenso , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/fisiologia , Células Eritroides/citologia , Células Eritroides/metabolismo , Inativação Gênica , Humanos , Células K562 , Camundongos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/fisiologia , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , gama-Globinas/metabolismo
18.
J Transl Med ; 11: 14, 2013 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-23316703

RESUMO

BACKGROUND: Pharmacologic reactivation of fetal hemoglobin expression is a promising strategy for treatment of sickle cell disease and ß-thalassemia. The objective of this study was to investigate the effect of the methyl transferase inhibitor adenosine-2',3'-dialdehyde (Adox) on induction of human fetal hemoglobin (HbF) in K562 cells and human hematopoietic progenitor cells. METHODS: Expression levels of human fetal hemoglobin were assessed by northern blot analysis and Real-time PCR. HbF and adult hemoglobin (HbA) content were analyzed using high-performance liquid chromatography (HPLC). DNA methylation levels on human gamma-globin gene promoters were determined using Bisulfite sequence analysis. Enrichment of histone marks on genes was assessed by chromosome immunoprecipitation (ChIP). RESULTS: Adox induced γ-globin gene expression in both K562 cells and in human bone marrow erythroid progenitor cells through a mechanism potentially involving inhibition of protein arginine methyltransferase 5 (PRMT5). CONCLUSIONS: The ability of methyl transferase inhibitors such as Adox to efficiently reactivate fetal hemoglobin expression suggests that these agents may provide a means of reactivating fetal globin expression as a therapeutic option for treating sickle cell disease and ß-thalassemia.


Assuntos
Adenosina/análogos & derivados , Hemoglobina Fetal/biossíntese , Adenosina/farmacologia , Northern Blotting , Imunoprecipitação da Cromatina , Cromatografia Líquida de Alta Pressão , Hemoglobina Fetal/genética , Humanos , Células K562 , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase em Tempo Real
19.
Haematologica ; 97(11): 1632-40, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22689669

RESUMO

BACKGROUND: Histone H3 lysine 4 (K4) methylation has been linked with transcriptional activity in mammalian cells. The WD40-repeat protein, WDR5, is an essential component of the MLL complex that induces histone H3 K4 methylation, but the role of WDR5 in human globin gene regulation has not yet been established. DESIGN AND METHODS: To study the role of WDR5 in human globin gene regulation, we performed knockdown experiments in both K562 cells and primary human bone marrow erythroid progenitor cells (BMC). The effects of WDR5 knockdown on γ-globin gene expression were determined. Biochemical approaches were also employed to investigate WDR5 interaction molecules. Chromosomal marks in the globin locus were analyzed by ChIP. RESULTS: We found that WDR5 interacted with protein arginine methyltransferase 5 (PRMT5), a known repressor of γ-globin gene expression, and was essential for generating tri-methylated H3K4 (H3K4me3) at the γ-globin promoter in K562 cells. Enforced expression of WDR5 in K562 cells reduced γ-globin gene expression, whereas knockdown of WDR5 increased γ-globin gene expression in both K562 cells and primary human bone marrow erythroid progenitor cells. Consistent with this, both histone H3 and H4 acetylation at the γ-globin promoter were increased, while histone H4R3 and H3K9 methylation were decreased, in WDR5 knockdown cells compared to controls. We found that WDR5 interacted with HDAC1 and a PHD domaincontaining protein, ING2 (inhibitor of growth), an H3K4me3 mark reader, to enhance γ-globin gene transcriptional repression. In human BMC, levels of WDR5 were highly enriched on the γ-promoter relative to levels on other globin promoters and compared to the γ-promoter in cord blood erythroid progenitors, suggesting that WDR5 is important in the developmental globin gene expression program. CONCLUSIONS: Our data are consistent with a model in which WDR5 binds the γ-globin promoter in a PRMT5-dependent manner; H3K4me3 induced at the γ-globin promoter by WDR5 may result in the recruitment of the ING2-associated HDAC1 component and consequent silencing of γ-globin gene expression.


Assuntos
Células Precursoras Eritroides/metabolismo , Globulinas Fetais/biossíntese , Inativação Gênica/fisiologia , Histona-Lisina N-Metiltransferase/metabolismo , Modelos Biológicos , Regiões Promotoras Genéticas/fisiologia , Células Precursoras Eritroides/citologia , Feminino , Globulinas Fetais/genética , Técnicas de Silenciamento de Genes , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Células K562 , Masculino , Metilação , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
20.
PLoS One ; 7(4): e34327, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22523547

RESUMO

SRY is a sex-determining gene that encodes a transcription factor, which triggers male development in most mammals. The molecular mechanism of SRY action in testis determination is, however, poorly understood. In this study, we demonstrate that WDR5, which encodes a WD-40 repeat protein, is a direct target of SRY. EMSA experiments and ChIP assays showed that SRY could bind to the WDR5 gene promoter directly. Overexpression of SRY in LNCaP cells significantly increased WDR5 expression concurrent with histone H3K4 methylation on the WDR5 promoter. To specifically address whether SRY contributes to WDR5 regulation, we introduced a 4-hydroxy-tamoxifen-inducible SRY allele into LNCaP cells. Conditional SRY expression triggered enrichment of SRY on the WDR5 promoter resulting in induction of WDR5 transcription. We found that WDR5 was self regulating through a positive feedback loop. WDR5 and SRY interacted and were colocalized in cells. In addition, the interaction of WDR5 with SRY resulted in activation of Sox9 while repressing the expression of ß-catenin. These results suggest that, in conjunction with SRY, WDR5 plays an important role in sex determination.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Fatores de Transcrição SOX9/biossíntese , Proteína da Região Y Determinante do Sexo/metabolismo , Testículo/embriologia , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Histona-Lisina N-Metiltransferase/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Regiões Promotoras Genéticas , Neoplasias da Próstata , Proteínas/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Testículo/metabolismo , beta Catenina/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...