Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Transl Med ; 22(1): 170, 2024 02 18.
Article in English | MEDLINE | ID: mdl-38368381

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is characterized by aggressive progression and elevated mortality rates. This study aimed to investigate the regulatory effects of RBBP7 on HCC pathogenesis and the underlying mechanisms. METHODS: The expression and clinical feature of RBBP7 were evaluated using bioinformatics analysis and the assessment of clinical HCC samples. CCK8 and colony formation were employed to estimate cell proliferation function of RBBP7. Aerobic glycolysis levels of RBBP7 were evaluated by measuring ATP levels, lactic acid production, glucose uptake capacity, and the expression of relevant enzymes (PFKM, PKM2, and LDHA). The phosphorylation levels in PI3K/AKT signaling were measured by western blotting. The regulatory effect of transcription factors of specificity protein 1 (SP1) on RBBP7 mRNA expression was confirmed in dual-luciferase reporter assays and chromatin immunoprecipitation experiments. The proliferation- and glycolysis-associated proteins were assessed using immunofluorescence staining in vivo. RESULTS: We found that RBBP7 is expressed at high levels in HCC and predicts poor survival. Functional assays showed that RBBP7 promoted HCC proliferation and glycolysis. Mechanistically, it was demonstrated that RBBP7 activates the PI3K/AKT pathway, a crucial pathway in glycolysis, contributing to the progression of HCC. The outcomes of the dual-luciferase assay further confirmed that SP1 is capable of activating the promoter of RBBP7. CONCLUSIONS: RBBP7, which is up-regulated by SP1, promotes HCC cell proliferation and glycolysis through the PI3K/AKT pathway. The findings of this study suggest that RBBP7 is a potential biomarker for HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , Luciferases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Retinoblastoma-Binding Protein 7/genetics , Retinoblastoma-Binding Protein 7/metabolism
2.
J Clin Invest ; 133(20)2023 10 16.
Article in English | MEDLINE | ID: mdl-37843278

ABSTRACT

Maturation arrest (MA) is a subtype of non-obstructive azoospermia, and male infertility is a known risk factor for testicular tumors. However, the genetic basis for many affected individuals remains unknown. Here, we identified a deleterious hemizygous variant of X-linked retinoblastoma-binding protein 7 (RBBP7) as a potential key cause of MA, which was also found to be associated with the development of Leydig cell tumors. This mutation resulted in premature protein translation termination, affecting the sixth WD40 domain of the RBBP7 and the interaction of the mutated RBBP7 with histone H4. Decreased BRCA1 and increased γH2AX were observed in the proband. In mouse spermatogonial and pachytene spermatocyte-derived cells, deprivation of rbbp7 led to cell cycle arrest and apoptosis. In Drosophila, knockdown of RBBP7/Caf1-55 in germ cells resulted in complete absence of germ cells and reduced testis size, whereas knockdown of RBBP7/Caf1-55 in cyst cells resulted in hyperproliferative testicular cells. Interestingly, male infertility caused by Caf1-55 deficiency was rescued by ectopic expression of wild-type human RBBP7 but not mutant variants, suggesting the importance of RBBP7 in spermatogenesis. Our study provides insights into the mechanisms underlying the co-occurrence of MA and testicular tumors and may pave the way for innovative genetic diagnostics of these 2 diseases.


Subject(s)
Azoospermia , Infertility, Male , Testicular Neoplasms , Animals , Humans , Male , Mice , Azoospermia/genetics , Azoospermia/metabolism , Azoospermia/pathology , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Mutation , Retinoblastoma-Binding Protein 7/genetics , Retinoblastoma-Binding Protein 7/metabolism , Spermatogenesis/genetics , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Testis/metabolism
3.
Cancer Gene Ther ; 30(8): 1124-1133, 2023 08.
Article in English | MEDLINE | ID: mdl-37225855

ABSTRACT

Colorectal cancer (CRC) poses one of the most serious threats to human health worldwide, and abnormally expressed c-Myc and p53 are deemed the pivotal driving forces of CRC progression. In this study, we discovered that the lncRNA FIT, which was downregulated in CRC clinical samples, was transcriptionally suppressed by c-Myc in vitro and promoted CRC cell apoptosis by inducing FAS expression. FAS is a p53 target gene, and we found that FIT formed a trimer with RBBP7 and p53 that facilitated p53 acetylation and p53-mediated FAS gene transcription. Moreover, FIT was capable of retarding CRC growth in a mouse xenograft model, and FIT expression was positively correlated with FAS expression in clinical samples. Thus, our study elucidates the role of the lncRNA FIT in human colorectal cancer growth and provides a potential target for anti-CRC drugs.


Subject(s)
Colorectal Neoplasms , RNA, Long Noncoding , Humans , Animals , Mice , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Acetylation , RNA, Long Noncoding/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Retinoblastoma-Binding Protein 7/genetics , Retinoblastoma-Binding Protein 7/metabolism
4.
Acta Biochim Biophys Sin (Shanghai) ; 54(2): 179-186, 2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35538026

ABSTRACT

Hypoxia-induced epigenetic regulation calls for more effective therapeutic targets for esophageal cancer. We used GEPIA and UALCAN databases to screen survival-related and cancer stage-associated genes. Eca109 and KYSE450 esophageal cancer cell lines were cultured under normoxia, hypoxia, or CoCl-induced hypoxia conditions, which were further transfected with plasmids expressing RB binding protein 7 (RBBP7), hypoxia-inducible factor 1 (HIF1)-α, or RBBP7 shRNA. Colony formation and MTT assays were used to detect cell proliferation. Tumor sphere formation and stemness marker detection were applied to assess cell stemness. RT-PCR and western blot analysis were used to detect the relative mRNA level and protein expression, respectively. Luciferase assay was utilized to detect the direct interaction between HIF1α and RBBP7. Up-regulated RBBP7 was identified as one of the most prominent survival-related genes, which is negatively correlated with the overall survival (OS), disease recurrence-free survival (DFS), and tumor stages. Hypoxia-induced HIF1α up-regulates RBBP7 expression, which promotes esophagus cancer cell viability, proliferation, and stemness with increased cyclin-dependent kinase 4 (CDK4) expression. Luciferase reporter assay verified that HIF1α transcriptionally regulates the expression of RBBP7. We conclude that hypoxia induces high expression of RBBP7 which is at least partially mediated by HIF1α, up-regulates the expression of downstream CDK4, and thereby promotes tumor progression in esophageal cancer cells.


Subject(s)
Cyclin-Dependent Kinase 4 , Esophageal Neoplasms , Retinoblastoma-Binding Protein 7 , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin-Dependent Kinase 4/biosynthesis , Cyclin-Dependent Kinase 4/genetics , Disease Progression , Epigenesis, Genetic , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Retinoblastoma-Binding Protein 7/biosynthesis , Retinoblastoma-Binding Protein 7/genetics , Retinoblastoma-Binding Protein 7/metabolism
5.
Epigenetics ; 17(10): 1205-1218, 2022 10.
Article in English | MEDLINE | ID: mdl-34709113

ABSTRACT

Preimplantation development is critical for reproductive successes in mammals. Thus, it is important to understand how preimplantation embryogenesis is regulated. As a key event of preimplantation development, epigenetic reprogramming has been widely studied, yet how epigenetic complexes regulate preimplantation development remains largely unknown. Retinoblastoma binding protein 4 (RBBP4) and 7 (RBBP7) are integral components of epigenetic complexes including SIN3A, NuRD, and CoREST. Here, we demonstrate that double knockdown of Rbbp4 and 7, but not individually, causes embryonic lethality during the morula-to-blastocyst transition. Mechanistically, depletion of RBBP4 and 7 results in dysregulation of genes related to cell cycle, lineage development, and regulation of transcription, which is accompanied by cell cycle block, disrupted lineage specification and chromatin structure. Interestingly, RBBP4/7 depletion leads to a dramatic increase in H3.3 and H3K27ac abundance during morula-to-blastocyst transition. ChIP-seq analysis in early embryos and embryonic stem cells reveals enrichment of H3.3 at the promoter regions of RBBP4/7 target genes. In summary, our studies demonstrate the compensatory role of RBBP4/7 and reveal its potential mechanisms in preimplantation development.Summary sentence:RBBP4 and RBBP7 play a compensatory role in regulating cell proliferation, apoptosis, and histone H3.3 deposition during preimplantation development.


Subject(s)
Histones , Retinoblastoma-Binding Protein 4 , Animals , Blastocyst/metabolism , Cell Proliferation , Chromatin/metabolism , DNA Methylation , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Histones/genetics , Histones/metabolism , Mammals/genetics , Mammals/metabolism , Mice , Retinoblastoma-Binding Protein 4/chemistry , Retinoblastoma-Binding Protein 4/genetics , Retinoblastoma-Binding Protein 4/metabolism , Retinoblastoma-Binding Protein 7/genetics , Retinoblastoma-Binding Protein 7/metabolism , Transcription Factors/genetics
6.
Nucleic Acids Res ; 49(11): 6196-6212, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34086947

ABSTRACT

Retinoblastoma-binding proteins 4 and 7 (RBBP4 and RBBP7) are two highly homologous human histone chaperones. They function in epigenetic regulation as subunits of multiple chromatin-related complexes and have been implicated in numerous cancers. Due to their overlapping functions, our understanding of RBBP4 and 7, particularly outside of Opisthokonts, has remained limited. Here, we report that in the ciliate protozoan Tetrahymena thermophila a single orthologue of human RBBP4 and 7 proteins, RebL1, physically interacts with histone H4 and functions in multiple epigenetic regulatory pathways. Functional proteomics identified conserved functional links for Tetrahymena RebL1 protein as well as human RBBP4 and 7. We found that putative subunits of multiple chromatin-related complexes including CAF1, Hat1, Rpd3, and MuvB, co-purified with RebL1 during Tetrahymena growth and conjugation. Iterative proteomics analyses revealed that the cell cycle regulatory MuvB-complex in Tetrahymena is composed of at least five subunits including evolutionarily conserved Lin54, Lin9 and RebL1 proteins. Genome-wide analyses indicated that RebL1 and Lin54 (Anqa1) bind within genic and intergenic regions. Moreover, Anqa1 targets primarily promoter regions suggesting a role for Tetrahymena MuvB in transcription regulation. RebL1 depletion inhibited cellular growth and reduced the expression levels of Anqa1 and Lin9. Consistent with observations in glioblastoma tumors, RebL1 depletion suppressed DNA repair protein Rad51 in Tetrahymena, thus underscoring the evolutionarily conserved functions of RBBP4/7 proteins. Our results suggest the essentiality of RebL1 functions in multiple epigenetic regulatory complexes in which it impacts transcription regulation and cellular viability.


Subject(s)
Histone Chaperones/metabolism , Protozoan Proteins/metabolism , Tetrahymena thermophila/metabolism , Amino Acid Sequence , Bacterial Proteins/metabolism , Biological Evolution , Conserved Sequence , DNA/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Expression , HEK293 Cells , Histone Chaperones/chemistry , Histone Chaperones/physiology , Histones/metabolism , Humans , Neoplasms/metabolism , Neoplasms/mortality , Oncogenes , Protozoan Proteins/chemistry , Protozoan Proteins/physiology , Retinoblastoma-Binding Protein 4/metabolism , Retinoblastoma-Binding Protein 7/metabolism , Tetrahymena thermophila/genetics , Tetrahymena thermophila/growth & development
7.
Acta Neuropathol ; 142(2): 279-294, 2021 08.
Article in English | MEDLINE | ID: mdl-33978814

ABSTRACT

Evidence indicates that tau hyper-phosphorylation and subsequent neurofibrillary tangle formation contribute to the extensive neuronal death in Alzheimer's disease (AD) and related tauopathies. Recent work has identified that increased tau acetylation can promote tau phosphorylation. Tau acetylation occurs at lysine 280 resulting from increased expression of the lysine acetyltransferase p300. The exact upstream mechanisms mediating p300 expression remain elusive. Additional work highlights the role of the epigenome in tau pathogenesis, suggesting that dysregulation of epigenetic proteins may contribute to acetylation and hyper-phosphorylation of tau. Here, we identify and focus on the histone-binding subunit of the Nucleosome Remodeling and Deacetylase (NuRD) complex: Retinoblastoma-Binding Protein 7 (Rbbp7). Rbbp7 chaperones chromatin remodeling proteins to their nuclear histone substrates, including histone acetylases and deacetylases. Notably, Rbbp7 binds to p300, suggesting that it may play a role in modulating tau acetylation. We interrogated Rbbp7 in post-mortem brain tissue, cell lines and mouse models of AD. We found reduced Rbbp7 mRNA expression in AD cases, a significant negative correlation with CERAD (neuritic plaque density) and Braak Staging (pathogenic tau inclusions) and a significant positive correlation with post-mortem brain weight. We also found a neuron-specific downregulation of Rbbp7 mRNA in AD patients. Rbbp7 protein levels were significantly decreased in 3xTg-AD and PS19 mice compared to NonTg, but no decreases were found in APP/PS1 mice that lack tau pathology. In vitro, Rbbp7 overexpression rescued TauP301L-induced cytotoxicity in immortalized hippocampal cells and primary cortical neurons. In vivo, hippocampal Rbbp7 overexpression rescued neuronal death in the CA1 of PS19 mice. Mechanistically, we found that increased Rbbp7 reduced p300 levels, tau acetylation at lysine 280 and tau phosphorylation at AT8 and AT100 sites. Collectively, these data identify a novel role of Rbbp7, protecting against tau-related pathologies, and highlight its potential as a therapeutic target in AD and related tauopathies.


Subject(s)
Acetylation , Neurons/pathology , Retinoblastoma-Binding Protein 7/metabolism , Tauopathies/pathology , tau Proteins/metabolism , Alzheimer Disease/pathology , Animals , Brain/pathology , Disease Models, Animal , Mice , Neurons/metabolism , Protein Processing, Post-Translational/physiology , Retinoblastoma-Binding Protein 7/genetics
8.
Nat Chem Biol ; 16(11): 1199-1207, 2020 11.
Article in English | MEDLINE | ID: mdl-32747809

ABSTRACT

Targeted protein degradation is a new therapeutic modality based on drugs that destabilize proteins by inducing their proximity to E3 ubiquitin ligases. Of particular interest are molecular glues that can degrade otherwise unligandable proteins by orchestrating direct interactions between target and ligase. However, their discovery has so far been serendipitous, thus hampering broad translational efforts. Here, we describe a scalable strategy toward glue degrader discovery that is based on chemical screening in hyponeddylated cells coupled to a multi-omics target deconvolution campaign. This approach led us to identify compounds that induce ubiquitination and degradation of cyclin K by prompting an interaction of CDK12-cyclin K with a CRL4B ligase complex. Notably, this interaction is independent of a dedicated substrate receptor, thus functionally segregating this mechanism from all described degraders. Collectively, our data outline a versatile and broadly applicable strategy to identify degraders with nonobvious mechanisms and thus empower future drug discovery efforts.


Subject(s)
Acetamides/chemistry , Anti-Bacterial Agents/pharmacology , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Doxycycline/pharmacology , Hydrazines/chemistry , Indoles/chemistry , Proteolysis/drug effects , Retinoblastoma-Binding Protein 7/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Gene Expression Regulation , Humans , Molecular Structure , Protein Binding , Protein Conformation , Protein Processing, Post-Translational/drug effects , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
9.
J Reprod Dev ; 66(2): 125-133, 2020 Apr 10.
Article in English | MEDLINE | ID: mdl-31956172

ABSTRACT

Kisspeptin, encoded by Kiss1, is essential for reproduction in mammals. Kiss1 expression is regulated by estrogen via histone acetylation in the Kiss1 promotor region. Thus, elucidation of histone modification factor(s) involved in the regulation of Kiss1 expression is required to gain further understanding of the mechanisms of its control. The RNA-seq analysis of isolated kisspeptin neurons, obtained from the arcuate nucleus (ARC) of female rats, revealed that Rbbp7, encoding retinoblastoma binding protein 7 (RBBP7), a member of histone modification and chromatin remodeling complexes, is highly expressed in the ARC kisspeptin neurons. Thus, the present study aimed to investigate whether RBBP7 is involved in Kiss1 expression. Histological analysis using in situ hybridization (ISH) revealed that Rbbp7 expression was located in several hypothalamic nuclei, including the ARC and the anteroventral periventricular nucleus (AVPV), where kisspeptin neurons are located. Double ISH for Rbbp7 and Kiss1 showed that a majority of kisspeptin neurons (more than 85%) expressed Rbbp7 mRNA in both the ARC and the AVPV of female rats. Further, Rbbp7 mRNA knockdown significantly decreased in vitro expression of Kiss1 in a mouse immortalized kisspeptin neuronal cell line (mHypoA-55). Estrogen treatment significantly decreased and increased Kiss1 mRNA levels in the ARC and AVPV of ovariectomized female rats, respectively, but failed to affect Rbbp7 mRNA levels in both the nuclei. Taken together, these findings suggest that RBBP7 is involved in the upregulation of Kiss1 expression in kisspeptin neurons of rodents in an estrogen-independent manner.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Hypothalamus, Anterior/metabolism , Kisspeptins/metabolism , Retinoblastoma-Binding Protein 7/metabolism , Up-Regulation , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Cell Line , Estradiol/pharmacology , Female , Hypothalamus, Anterior/drug effects , Kisspeptins/genetics , Neurons/drug effects , Neurons/metabolism , Rats , Retinoblastoma-Binding Protein 7/genetics
10.
Eur Rev Med Pharmacol Sci ; 24(1): 151-163, 2020 01.
Article in English | MEDLINE | ID: mdl-31957828

ABSTRACT

OBJECTIVE: Esophageal cancer (EC) ranks as the sixth leading cause of cancer-related mortality worldwide. Circular RNAs (circRNAs) are involved in the pathogenesis of different cancers. However, the regulatory mechanism of circ_0006168 in EC progression is still unclear. MATERIALS AND METHODS: The expression of circ_0006168, microRNA (miR)-384, and retinoblastoma binding protein 7 (RBBP7) in tumors and cells was measured by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). The stability of circ_0006168 was analyzed after RNase R treatment. 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay was conducted to evaluate cell viability. Transwell assay was applied to determine cell migration and invasion. Glucose consumption and lactate production were detected using glucose detection and lactic acid detection kits. The interaction between miR-384 and circ_0006168 or RBBP7 was certified by Dual-Luciferase reporter system. Protein expression of pyruvate kinase (PK), RBBP7, S6 ribosomal protein kinase (S6K), phosphorylated S6K (p-S6K), S6, phosphorylated S6 (p-S6) was analyzed by Western blot. RESULTS: Circ_0006168 and RBBP7 were over-expressed while miR-384 was low-expressed in EC tumors and cells. The repression of circ_0006168 attenuated cell proliferation, migration, invasion, and glycolysis in EC. Of note, circ_0006168 functioned as a sponge while RBBP7 acted as a target of miR-384 in EC. Rescue experiment revealed that miR-384 inhibitor abrogated circ_0006168 silencing-induced repression on cell proliferation, migration, and invasion in EC. Meanwhile, upregulation of RBBP7 restored the inhibition of miR-384 on EC cell progression. Moreover, circ_0006168 was able to improve RBBP7 level by interacting with miR-384. Also, circ_0006168 could activate S6K/S6 pathway by regulating RBBP7 expression. CONCLUSIONS: Abundance of circ_0006168 contributes to cell proliferation, migration, invasion, and glycolysis in EC by competitively sponging miR-384 to facilitate RBBP7 expression, representing prospective targets for EC therapy.


Subject(s)
Esophageal Neoplasms/metabolism , MicroRNAs/metabolism , RNA, Circular/metabolism , Retinoblastoma-Binding Protein 7/metabolism , Ribosomal Protein S6 Kinases/metabolism , Ribosomal Protein S6/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Humans , MicroRNAs/genetics , RNA, Circular/genetics , Retinoblastoma-Binding Protein 7/genetics , Ribosomal Protein S6/genetics , Ribosomal Protein S6 Kinases/genetics
11.
Nat Commun ; 11(1): 24, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31911655

ABSTRACT

The spindle assembly checkpoint (SAC) prevents premature chromosome segregation by inactivating the anaphase promoting complex/cyclosome (APC/C) until all chromosomes are properly attached to mitotic spindles. Here we identify a role for Cullin-RING ubiquitin ligase complex 4 (CRL4), known for modulating DNA replication, as a crucial mitotic regulator that triggers the termination of the SAC and enables chromosome segregation. CRL4 is recruited to chromatin by the replication origin binding protein RepID/DCAF14/PHIP. During mitosis, CRL4 dissociates from RepID and replaces it with RB Binding Protein 7 (RBBP7), which ubiquitinates the SAC mediator BUB3 to enable mitotic exit. During interphase, BUB3 is protected from CRL4-mediated degradation by associating with promyelocytic leukemia (PML) nuclear bodies, ensuring its availability upon mitotic onset. Deficiencies in RepID, CRL4 or RBBP7 delay mitotic exit, increase genomic instability and enhance sensitivity to paclitaxel, a microtubule stabilizer and anti-tumor drug.


Subject(s)
Anaphase , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Metaphase , Poly-ADP-Ribose Binding Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Cycle Proteins/genetics , Cell Line , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mitosis , Poly-ADP-Ribose Binding Proteins/genetics , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Protein Binding , Proteolysis , Retinoblastoma-Binding Protein 7/genetics , Retinoblastoma-Binding Protein 7/metabolism , Spindle Apparatus/metabolism , Ubiquitin-Protein Ligases/genetics
12.
Biochem Biophys Res Commun ; 501(2): 440-447, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29738775

ABSTRACT

The E3 ubiquitin ligase HUWE1/Mule/ARF-BP1 plays an important role in diverse biological processes including DNA damage repair and apoptosis. Our previous study has shown that in response to DNA damage HUWE1 was downregulated in CUL4B-mediated ubiquitination and subsequent proteasomal degradation, and CUL4B-mediated regulation of HUWE1 was important for cell survival upon DNA damage. CUL4B is a core component of the CUL4B Ring ligase complexes containing ROC1, DDB1 and a DDB1-Cullin Associated Factors (DCAFs), the latter of which are DDB1-binding WD40 adaptors critical for substrate recognition and recruitment. However, the identity of DCAF in CRL4B that mediates degradation of HUWE1 remains elusive. Here we report that RBBP7 is the DCAF in the CRL4B complex bridging the DDB1-CUL4B-ROC1 to HUWE1. Loading of HUWE1 to the E3 ubiquitin ligase complex resulted in its polyubiquitination, and consequently its proteasome mediated degradation. Overexpression of RBBP7 promoted HUWE1 protein degradation, while depletion of RBBP7 stabilized HUWE1, and hence accelerated the degradation of MCL-1 and BRCA1, two substrates of HUWE1 that are critical in apoptosis and DNA damage repair. Taken together, these data reveal CRL4BRBBP7 is the E3 ligase responsible for the proteasomal degradation of HUWE1, and further provide a potential strategy for cancer therapy by targeting HUWE1 and the CUL4B E3 ligase complex.


Subject(s)
Retinoblastoma-Binding Protein 7/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , BRCA1 Protein/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cullin Proteins/genetics , Cullin Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Stability , Proteolysis , Retinoblastoma-Binding Protein 7/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
13.
Mol Oncol ; 12(4): 476-494, 2018 04.
Article in English | MEDLINE | ID: mdl-29377600

ABSTRACT

Cullin 4B, a member of the Cullins, which serve as scaffolds to facilitate the assembly of E3 ligase complexes, is aberrantly expressed in many cancers, including osteosarcoma. Recently, we observed that CUL4B forms the CRL4BDCAF11 E3 ligase, which specifically ubiquitinates and degrades the cyclin-dependent kinase (CDK) inhibitor p21Cip1 in human osteosarcoma cells. However, the underlying mechanisms regarding the aberrant expression of CUL4B and the upstream members of this signaling pathway are mostly unknown. In this study, we demonstrate that nuclear factor kappaB (NF-κB) is a direct modulator of CUL4B expression. The CUL4B promoter is responsive to several NF-κB subunits, including RelA, RelB, and c-Rel, but not to p50 or p52. Additional studies reveal that the tumor necrosis factor alpha (TNF-α)/NF-κB axis pathway is activated in human osteosarcoma cells. This activation causes both CUL4B and NF-κB subunits to become abundant in the nucleus of human osteosarcoma cells. The down-regulation of individual genes, including TNFR1, RelA, RelB, c-Rel, and CUL4B, or pairs of them, including TNFR1 + RelA, TNFR1 + RelB, TNFR1 + c-Rel, and RelA+CUL4B, has similar effects on cell growth inhibition, colony formation, cell invasion, and in vivo tumor formation, whereas the overexpression of CUL4B in these knockdown cells significantly reverses their phenotypes. The inhibition of the TNF-α/NF-κB pathway greatly attenuates CRL4BDCAF11 E3 ligase activity and causes the accumulation of p21Cip1 , thereby leading to cell cycle arrest at the S phase. Taken together, our results support a model in which the activation of the TNF-α/NF-κB axis contributes to an increase in CRL4BDCAF11 activity and a decrease in p21Cip1 protein levels, thereby controlling cell cycle progression in human osteosarcoma cells.


Subject(s)
Bone Neoplasms/metabolism , Carrier Proteins/metabolism , Cell Cycle , Models, Biological , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Osteosarcoma/metabolism , Retinoblastoma-Binding Protein 7/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Female , Humans , Male , Mice , Mice, Nude , NF-kappa B/genetics , Neoplasm Proteins/genetics , Osteosarcoma/genetics , Osteosarcoma/pathology , Retinoblastoma-Binding Protein 7/genetics , Tumor Necrosis Factor-alpha/genetics , Ubiquitin-Protein Ligase Complexes
14.
J Biol Chem ; 293(6): 2125-2136, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29263092

ABSTRACT

The transcription factor BCL11A has recently been reported to be a driving force in triple-negative breast cancer (TNBC), contributing to the maintenance of a chemoresistant breast cancer stem cell (BCSC) population. Although BCL11A was shown to suppress γ-globin and p21 and to induce MDM2 expression in the hematopoietic system, its downstream targets in TNBC are still unclear. For its role in transcriptional repression, BCL11A was found to interact with several corepressor complexes; however, the mechanisms underlying these interactions remain unknown. Here, we reveal that BCL11A interacts with histone methyltransferase (PRC2) and histone deacetylase (NuRD and SIN3A) complexes through their common subunit, RBBP4/7. In fluorescence polarization assays, we show that BCL11A competes with histone H3 for binding to the negatively charged top face of RBBP4. To define that interaction, we solved the crystal structure of RBBP4 in complex with an N-terminal peptide of BCL11A (residues 2-16, BCL11A(2-16)). The crystal structure identifies novel interactions between BCL11A and the side of the ß-propeller of RBBP4 that are not seen with histone H3. We next show that BCL11A(2-16) pulls down RBBP4, RBBP7, and other components of PRC2, NuRD, and SIN3A from the cell lysate of the TNBC cell line SUM149. Furthermore, we demonstrate the therapeutic potential of targeting the RBBP4-BCL11A binding by showing that a BCL11A peptide can decrease aldehyde dehydrogenase-positive BCSCs and mammosphere formation capacity in SUM149. Together, our findings have uncovered a previously unidentified mechanism that BCL11A may use to recruit epigenetic complexes to regulate transcription and promote tumorigenesis.


Subject(s)
Carrier Proteins/metabolism , Nuclear Proteins/metabolism , Retinoblastoma-Binding Protein 4/metabolism , Retinoblastoma-Binding Protein 7/metabolism , Carcinogenesis , Carrier Proteins/chemistry , Cell Line , Crystallography, X-Ray , Epigenomics , Histone Deacetylases/metabolism , Histone Methyltransferases/metabolism , Humans , Nuclear Proteins/chemistry , Protein Binding , Repressor Proteins , Retinoblastoma-Binding Protein 4/chemistry , Retinoblastoma-Binding Protein 7/chemistry , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/pathology
15.
Sci Signal ; 10(464)2017 01 31.
Article in English | MEDLINE | ID: mdl-28143904

ABSTRACT

Adenosine monophosphate (AMP)-activated protein kinase (AMPK) acts as a master regulator of cellular energy homeostasis by directly phosphorylating metabolic enzymes and nutrient transporters and by indirectly promoting the transactivation of nuclear genes involved in mitochondrial biogenesis and function. We explored the mechanism of AMPK-mediated induction of gene expression. We identified AMPK consensus phosphorylation sequences in three proteins involved in nucleosome remodeling: DNA methyltransferase 1 (DNMT1), retinoblastoma binding protein 7 (RBBP7), and histone acetyltransferase 1 (HAT1). DNMT1 mediates DNA methylation that limits transcription factor access to promoters and is inhibited by RBBP7. Acetylation of histones by HAT1 creates a more relaxed chromatin-DNA structure that favors transcription. AMPK-mediated phosphorylation resulted in the activation of HAT1 and inhibition of DNMT1. For DNMT1, this inhibition was both a direct effect of phosphorylation and the result of increased interaction with RBBP7. In human umbilical vein cells, pharmacological AMPK activation or pulsatile shear stress triggered nucleosome remodeling and decreased cytosine methylation, leading to increased expression of nuclear genes encoding factors involved in mitochondrial biogenesis and function, such as peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), transcription factor A (Tfam), and uncoupling proteins 2 and 3 (UCP2 and UCP3). Similar effects were seen in the aortas of mice given pharmacological AMPK activators, and these effects required AMPK2α. These results enhance our understanding of AMPK-mediated mitochondrial gene expression through nucleosome remodeling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Histone Acetyltransferases/metabolism , Organelle Biogenesis , Retinoblastoma-Binding Protein 7/metabolism , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Acetyltransferases/genetics , Humans , Immunoblotting , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Retinoblastoma-Binding Protein 7/genetics , Tandem Mass Spectrometry , Transcription Factors/genetics , Transcription Factors/metabolism , Uncoupling Protein 2/genetics , Uncoupling Protein 2/metabolism , Uncoupling Protein 3/genetics , Uncoupling Protein 3/metabolism
16.
Nat Commun ; 7: 13465, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27811920

ABSTRACT

Centromeres are specified epigenetically through the deposition of the centromere-specific histone H3 variant CENP-A. However, how additional epigenetic features are involved in centromere specification is unknown. Here, we find that histone H4 Lys5 and Lys12 acetylation (H4K5ac and H4K12ac) primarily occur within the pre-nucleosomal CENP-A-H4-HJURP (CENP-A chaperone) complex, before centromere deposition. We show that H4K5ac and H4K12ac are mediated by the RbAp46/48-Hat1 complex and that RbAp48-deficient DT40 cells fail to recruit HJURP to centromeres and do not incorporate new CENP-A at centromeres. However, C-terminally-truncated HJURP, that does not bind CENP-A, does localize to centromeres in RbAp48-deficient cells. Acetylation-dead H4 mutations cause mis-localization of the CENP-A-H4 complex to non-centromeric chromatin. Crucially, CENP-A with acetylation-mimetic H4 was assembled specifically into centromeres even in RbAp48-deficient DT40 cells. We conclude that H4K5ac and H4K12ac, mediated by RbAp46/48, facilitates efficient CENP-A deposition into centromeres.


Subject(s)
Centromere Protein A/metabolism , Centromere/metabolism , Histones/metabolism , Molecular Chaperones/metabolism , Nucleosomes/metabolism , Acetylation , Animals , Cell Line, Tumor , Centromere/genetics , Centromere Protein A/genetics , Chickens , Chromatin/metabolism , Epigenesis, Genetic , Histones/genetics , Humans , Lysine/metabolism , Molecular Chaperones/genetics , Mutation , Nucleosomes/genetics , Retinoblastoma-Binding Protein 4/metabolism , Retinoblastoma-Binding Protein 7/metabolism
17.
Protein Sci ; 25(8): 1472-82, 2016 08.
Article in English | MEDLINE | ID: mdl-27144666

ABSTRACT

The nucleosome remodeling and deacetylase (NuRD) complex remodels the genome in the context of both gene transcription and DNA damage repair. It is essential for normal development and is distributed across multiple tissues in organisms ranging from mammals to nematode worms. In common with other chromatin-remodeling complexes, however, its molecular mechanism of action is not well understood and only limited structural information is available to show how the complex is assembled. As a step towards understanding the structure of the NuRD complex, we have characterized the interaction between two subunits: the metastasis associated protein MTA1 and the histone-binding protein RBBP4. We show that MTA1 can bind to two molecules of RBBP4 and present negative stain electron microscopy and chemical crosslinking data that allow us to build a low-resolution model of an MTA1-(RBBP4)2 subcomplex. These data build on our understanding of NuRD complex structure and move us closer towards an understanding of the biochemical basis for the activity of this complex.


Subject(s)
Histone Deacetylases/chemistry , Nucleosomes/chemistry , Protein Subunits/chemistry , Repressor Proteins/chemistry , Retinoblastoma-Binding Protein 4/chemistry , Retinoblastoma-Binding Protein 7/chemistry , Amino Acid Sequence , Animals , Cloning, Molecular , Conserved Sequence , Cross-Linking Reagents/chemistry , Gene Expression , HEK293 Cells , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Kinetics , Models, Molecular , Mutation , Nucleosomes/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Secondary , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retinoblastoma-Binding Protein 4/genetics , Retinoblastoma-Binding Protein 4/metabolism , Retinoblastoma-Binding Protein 7/genetics , Retinoblastoma-Binding Protein 7/metabolism , Sequence Alignment , Thermodynamics , Trans-Activators , Transcription, Genetic
18.
Mol Cell Biol ; 36(13): 1881-98, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27161321

ABSTRACT

The transcription factor BCL11B/CTIP2 is a major regulatory protein implicated in various aspects of development, function and survival of T cells. Mitogen-activated protein kinase (MAPK)-mediated phosphorylation and SUMOylation modulate BCL11B transcriptional activity, switching it from a repressor in naive murine thymocytes to a transcriptional activator in activated thymocytes. Here, we show that BCL11B interacts via its conserved N-terminal MSRRKQ motif with endogenous MTA1 and MTA3 proteins to recruit various NuRD complexes. Furthermore, we demonstrate that protein kinase C (PKC)-mediated phosphorylation of BCL11B Ser2 does not significantly impact BCL11B SUMOylation but negatively regulates NuRD recruitment by dampening the interaction with MTA1 or MTA3 (MTA1/3) and RbAp46 proteins. We detected increased phosphorylation of BCL11B Ser2 upon in vivo activation of transformed and primary human CD4(+) T cells. We show that following activation of CD4(+) T cells, BCL11B still binds to IL-2 and Id2 promoters but activates their transcription by recruiting P300 instead of MTA1. Prolonged stimulation results in the direct transcriptional repression of BCL11B by KLF4. Our results unveil Ser2 phosphorylation as a new BCL11B posttranslational modification linking PKC signaling pathway to T-cell receptor (TCR) activation and define a simple model for the functional switch of BCL11B from a transcriptional repressor to an activator during TCR activation of human CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Protein Kinase C/metabolism , Repressor Proteins/metabolism , Serine/metabolism , Tumor Suppressor Proteins/metabolism , HEK293 Cells , Histone Deacetylases/metabolism , Humans , Interleukin-2/metabolism , Jurkat Cells , Kruppel-Like Factor 4 , Lymphocyte Activation , Neoplasm Proteins/metabolism , Phosphorylation , Repressor Proteins/chemistry , Retinoblastoma-Binding Protein 7/metabolism , Trans-Activators , Tumor Suppressor Proteins/chemistry
19.
J Mol Neurosci ; 58(2): 243-53, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26553261

ABSTRACT

The liver X receptor agonist, GW3965, improves cognition in Alzheimer's disease (AD) mouse models. Here, we determined if short-term GW3965 treatment induces changes in the DNA methylation state of the hippocampus, which are associated with cognitive improvement. Twenty-four-month-old triple-transgenic AD (3xTg-AD) mice were treated with GW3965 (50 mg/kg/day for 6 days). DNA methylation state was examined by modified bisulfite conversion and hybridization on Illumina Infinium Methylation BeadChip 450 k arrays. The Morris water maze was used for behavioral analysis. Our results show in addition to improvement in cognition methylation changes in 39 of 13,715 interrogated probes in treated 3xTg-AD mice compared with untreated 3xTg-AD mice. These changes in methylation probes include 29 gene loci. Importantly, changes in methylation status were mainly from synapse-related genes (SYP, SYN1, and DLG3) and neurogenesis-associated genes (HMGB3 and RBBP7). Thus, our results indicate that liver X receptors (LXR) agonist treatment induces rapid changes in DNA methylation, particularly in loci associated with genes involved in neurogenesis and synaptic function. Our results suggest a new potential mechanism to explain the beneficial effect of GW3965.


Subject(s)
Alzheimer Disease/metabolism , Benzoates/pharmacology , Benzylamines/pharmacology , DNA Methylation/drug effects , Neurogenesis , Orphan Nuclear Receptors/agonists , Synapses/drug effects , Alzheimer Disease/genetics , Animals , Female , HMGB3 Protein/genetics , HMGB3 Protein/metabolism , Liver X Receptors , Mice , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Retinoblastoma-Binding Protein 7/genetics , Retinoblastoma-Binding Protein 7/metabolism , Synapses/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism
20.
Biol Reprod ; 93(1): 13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26040671

ABSTRACT

Uterine stromal cells undergo extensive proliferation and differentiation during postimplantation development, a process known as decidualization. While a range of signaling molecules have been demonstrated to play essential roles in this event, its potential epigenetic regulatory mechanisms remain largely unknown. Retinoblastoma binding protein 7 (Rbbp7) is a protein reported as a core component of many histone modification and chromatin remodeling complexes. In the present study, our in situ hybridization and immunochemistry analysis first reveals a spatiotemporal expression of Rbbp7 in the uterus during the peri-implantation period. Observations of remarkable induction of Rbbp7 expression in uterine stromal cells in response to progesterone-nuclear receptor PR signaling point to its potential physiological significance during postimplantation uterine development. Employing a stealth RNA knockdown approach, combined with primary murine uterine stromal cell culture and an in vitro-induced decidualization model, we further demonstrate that Rbbp7 silencing compromises stromal cell decidualization via attenuating histone H4 acetylation and cyclin D3 expression. The results collectively suggest that Rbbp7 is a potentially functional player regulating normal histone acetylation modification and cyclin D3 expression in stromal cells during postimplantation decidual development.


Subject(s)
Embryo Implantation/physiology , Retinoblastoma-Binding Protein 7/metabolism , Stromal Cells/metabolism , Uterus/metabolism , Acetylation , Animals , Cell Differentiation , Cell Proliferation , Cyclin D3/metabolism , Female , Mice , Retinoblastoma-Binding Protein 7/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...