Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Proc Natl Acad Sci U S A ; 121(22): e2401729121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38768345

ABSTRACT

O-GlcNAc transferase (OGT) is an essential mammalian enzyme that glycosylates myriad intracellular proteins and cleaves the transcriptional coregulator Host Cell Factor 1 to regulate cell cycle processes. Via these catalytic activities as well as noncatalytic protein-protein interactions, OGT maintains cell homeostasis. OGT's tetratricopeptide repeat (TPR) domain is important in substrate recognition, but there is little information on how changing the TPR domain impacts its cellular functions. Here, we investigate how altering OGT's TPR domain impacts cell growth after the endogenous enzyme is deleted. We find that disrupting the TPR residues required for OGT dimerization leads to faster cell growth, whereas truncating the TPR domain slows cell growth. We also find that OGT requires eight of its 13 TPRs to sustain cell viability. OGT-8, like the nonviable shorter OGT variants, is mislocalized and has reduced Ser/Thr glycosylation activity; moreover, its interactions with most of wild-type OGT's binding partners are broadly attenuated. Therefore, although OGT's five N-terminal TPRs are not essential for cell viability, they are required for proper subcellular localization and for mediating many of OGT's protein-protein interactions. Because the viable OGT truncation variant we have identified preserves OGT's essential functions, it may facilitate their identification.


Subject(s)
N-Acetylglucosaminyltransferases , N-Acetylglucosaminyltransferases/metabolism , N-Acetylglucosaminyltransferases/genetics , Humans , Tetratricopeptide Repeat , Glycosylation , Host Cell Factor C1/metabolism , Host Cell Factor C1/genetics , HEK293 Cells , Protein Domains , Cell Proliferation , Cell Survival , Animals , Protein Binding
2.
Mol Cell ; 81(12): 2611-2624.e10, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33857404

ABSTRACT

The Shieldin complex shields double-strand DNA breaks (DSBs) from nucleolytic resection. Curiously, the penultimate Shieldin component, SHLD1, is one of the least abundant mammalian proteins. Here, we report that the transcription factors THAP1, YY1, and HCF1 bind directly to the SHLD1 promoter, where they cooperatively maintain the low basal expression of SHLD1, thereby ensuring a proper balance between end protection and resection during DSB repair. The loss of THAP1-dependent SHLD1 expression confers cross-resistance to poly (ADP-ribose) polymerase (PARP) inhibitor and cisplatin in BRCA1-deficient cells and shorter progression-free survival in ovarian cancer patients. Moreover, the embryonic lethality and PARPi sensitivity of BRCA1-deficient mice is rescued by ablation of SHLD1. Our study uncovers a transcriptional network that directly controls DSB repair choice and suggests a potential link between DNA damage and pathogenic THAP1 mutations, found in patients with the neurodevelopmental movement disorder adult-onset torsion dystonia type 6.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Cycle Proteins/genetics , DNA/metabolism , DNA Breaks, Double-Stranded/drug effects , DNA End-Joining Repair/drug effects , DNA Repair/genetics , Dystonia/genetics , Female , Host Cell Factor C1/metabolism , Mad2 Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Recombinational DNA Repair/drug effects , Telomere-Binding Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , YY1 Transcription Factor/metabolism
3.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33419956

ABSTRACT

O-GlcNAc transferase (OGT), found in the nucleus and cytoplasm of all mammalian cell types, is essential for cell proliferation. Why OGT is required for cell growth is not known. OGT performs two enzymatic reactions in the same active site. In one, it glycosylates thousands of different proteins, and in the other, it proteolytically cleaves another essential protein involved in gene expression. Deconvoluting OGT's myriad cellular roles has been challenging because genetic deletion is lethal; complementation methods have not been established. Here, we developed approaches to replace endogenous OGT with separation-of-function variants to investigate the importance of OGT's enzymatic activities for cell viability. Using genetic complementation, we found that OGT's glycosyltransferase function is required for cell growth but its protease function is dispensable. We next used complementation to construct a cell line with degron-tagged wild-type OGT. When OGT was degraded to very low levels, cells stopped proliferating but remained viable. Adding back catalytically inactive OGT rescued growth. Therefore, OGT has an essential noncatalytic role that is necessary for cell proliferation. By developing a method to quantify how OGT's catalytic and noncatalytic activities affect protein abundance, we found that OGT's noncatalytic functions often affect different proteins from its catalytic functions. Proteins involved in oxidative phosphorylation and the actin cytoskeleton were especially impacted by the noncatalytic functions. We conclude that OGT integrates both catalytic and noncatalytic functions to control cell physiology.


Subject(s)
Cell Proliferation/genetics , Fibroblasts/metabolism , Host Cell Factor C1/genetics , N-Acetylglucosaminyltransferases/genetics , Animals , Fibroblasts/cytology , Gene Expression Profiling , Gene Expression Regulation , Gene Knockout Techniques , Gene Ontology , Genetic Complementation Test , Glycosylation , HEK293 Cells , Host Cell Factor C1/metabolism , Humans , Metabolic Networks and Pathways/genetics , Mice , Molecular Sequence Annotation , N-Acetylglucosaminyltransferases/deficiency , Proteolysis
4.
Elife ; 102021 01 08.
Article in English | MEDLINE | ID: mdl-33416496

ABSTRACT

The oncoprotein transcription factor MYC is a major driver of malignancy and a highly validated but challenging target for the development of anticancer therapies. Novel strategies to inhibit MYC may come from understanding the co-factors it uses to drive pro-tumorigenic gene expression programs, providing their role in MYC activity is understood. Here we interrogate how one MYC co-factor, host cell factor (HCF)-1, contributes to MYC activity in a human Burkitt lymphoma setting. We identify genes connected to mitochondrial function and ribosome biogenesis as direct MYC/HCF-1 targets and demonstrate how modulation of the MYC-HCF-1 interaction influences cell growth, metabolite profiles, global gene expression patterns, and tumor growth in vivo. This work defines HCF-1 as a critical MYC co-factor, places the MYC-HCF-1 interaction in biological context, and highlights HCF-1 as a focal point for development of novel anti-MYC therapies.


Tumours form when cells lose control of their growth. Usually, cells produce signals that control how much and how often they divide. But if these signals become faulty, cells may grow too quickly or multiply too often. For example, a group of proteins known as MYC proteins activate growth genes in a cell, but too much of these proteins causes cells to grow uncontrollably. With one third of all cancer deaths linked to excess MYC proteins, these molecules could be key targets for anti-cancer drugs. However, current treatments fail to target these proteins. One option for treating cancers linked to MYC proteins could be to target proteins that work alongside MYC proteins, such as the protein HCF-1, which can attach to MYC proteins. To test if HCF-1 could be a potential drug target, Popay et al. first studied how HCF-1 and MYC proteins interacted using specific cancer cells grown in the laboratory. This revealed that when the two proteins connected, they activated genes that trigger rapid cell growth. When these cancer cells were then injected into mice, tumours quickly grew. However, when the MYC and HCF-1 attachments in the cancer cells were disrupted, the tumours shrunk. This suggests that if anti-cancer drugs were able to target HCF-1 proteins, they could potentially reduce or even reverse the growth of tumours. While further research is needed to identify drug candidates, these findings reveal a promising target for treating tumours that stem from over-abundant MYC proteins.


Subject(s)
Gene Expression , Genes, Mitochondrial , Host Cell Factor C1/genetics , Organelle Biogenesis , Proto-Oncogene Proteins c-myc/genetics , Ribosomes/physiology , Animals , Burkitt Lymphoma , Female , Host Cell Factor C1/metabolism , Humans , Mice , Mice, Nude , Proto-Oncogene Proteins c-myc/metabolism
5.
Cell Death Dis ; 11(10): 907, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097698

ABSTRACT

The eukaryotic cell cycle involves a highly orchestrated series of events in which the cellular genome is replicated during a synthesis (S) phase and each of the two resulting copies are segregated properly during mitosis (M). Host cell factor-1 (HCF-1) is a transcriptional co-regulator that is essential for and has been implicated in basic cellular processes, such as transcriptional regulation and cell cycle progression. Although a series of HCF-1 transcriptional targets have been identified, few functional clues have been provided, especially for chromosome segregation. Our results showed that HCF-1 activated CDC42 expression by binding to the -881 to -575 region upstream of the CDC42 transcription start site, and the regulation of CDC42 expression by HCF-1 was correlated with cell cycle progression. The overexpression of a spontaneously cycling and constitutively active CDC42 mutant (CDC42F28L) rescued G1 phase delay and multinucleate defects in mitosis upon the loss of HCF-1. Therefore, these results establish that HCF-1 ensures proper cell cycle progression by regulating the expression of CDC42, which indicates a possible mechanism of cell cycle coordination and the regulation mode of typical Rho GTPases.


Subject(s)
Host Cell Factor C1/metabolism , cdc42 GTP-Binding Protein/metabolism , Cell Cycle/physiology , Chromosome Segregation , Cyclin A/biosynthesis , Cyclin A/genetics , Disease Progression , G1 Phase Cell Cycle Checkpoints , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Host Cell Factor C1/genetics , Humans , Mitosis , Promoter Regions, Genetic , cdc42 GTP-Binding Protein/biosynthesis , cdc42 GTP-Binding Protein/genetics
6.
BMB Rep ; 53(12): 634-639, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33050986

ABSTRACT

In prostate cancer, the androgen receptor (AR) transcription factor is a major regulator of cell proliferation and metastasis. To identify new AR regulators, we focused on Mixed lineage leukemia 5 (MLL5), a histone-regulating enzyme, because significantly higher MLL5 expression was detected in prostate cancer tissues than in matching normal tissues. When we expressed shRNAs targeting MLL5 gene in prostate cancer cell line, the growth rate and AR activity were reduced compared to those in control cells, and migration ability of the knockdown cells was reduced significantly. To determine the molecular mechanisms of MLL5 on AR activity, we proved that AR physically interacted with MLL5 and other co-factors, including SET-1 and HCF-1, using an immunoprecipitation method. The chromatin immunoprecipitation analysis showed reduced binding of MLL5, co-factors, and AR enzymes to AR target gene promoters in MLL5 shRNA-expressing cells. Histone H3K4 methylation on the AR target gene promoters was reduced, and H3K9 methylation at the same site was increased in MLL5 knockdown cells. Finally, xenograft tumor formation revealed that reduction of MLL5 in prostate cancer cells retarded tumor growth. Our results thus demonstrate the important role of MLL5 as a new epigenetic regulator of AR in prostate cancer. [BMB Reports 2020; 53(12): 634-639].


Subject(s)
DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Host Cell Factor C1/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Histone-Lysine N-Methyltransferase/physiology , Histones/metabolism , Host Cell Factor C1/physiology , Humans , Male , Methylation , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/metabolism , Protein Processing, Post-Translational/genetics , Receptors, Androgen/metabolism , Receptors, Androgen/physiology , Signal Transduction/genetics , Transcription Factors/metabolism
7.
Nucleic Acids Res ; 48(13): 7321-7332, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32479599

ABSTRACT

The Siah1 and Siah2 ubiquitin ligases are implicated in diverse biological processes ranging from cellular stress responses, signaling to transcriptional regulation. A key substrate of Siah1 is ELL2, which undergoes proteolysis upon polyubiquitination. ELL2 stimulates transcriptional elongation and is a subunit of the Super Elongation Complex (SEC) essential for HIV-1 transactivation. Previously, multiple transcriptional and post-translational mechanisms are reported to control Siah's expression and activity. Here we show that the activity of Siah1/2 can also be suppressed by host cell factor 1 (HCF1), and the hitherto poorly characterized HCF2, which themselves are not degraded but can bind and block the substrate-binding domain (SBD) of Siah1/2 to prevent their autoubiquitination and trans-ubiquitination of downstream targets including ELL2. This effect stabilizes ELL2 and enhances the ELL2-SEC formation for robust HIV-1 transactivation. Thus, our study not only identifies HCF1/2 as novel activators of HIV-1 transcription through inhibiting Siah1 to stabilize ELL2, but also reveals the SBD of Siah1/2 as a previously unrecognized new target for HCF1/2 to exert this inhibition.


Subject(s)
Host Cell Factor C1/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Binding Sites , Cell Line , Chlorocebus aethiops , HeLa Cells , Humans , Nuclear Proteins/chemistry , Protein Binding , Transcriptional Elongation Factors/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitination
8.
Cell Rep ; 29(6): 1645-1659.e9, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31693902

ABSTRACT

Molecular chaperones such as heat-shock proteins (HSPs) help in protein folding. Their function in the cytosol has been well studied. Notably, chaperones are also present in the nucleus, a compartment where proteins enter after completing de novo folding in the cytosol, and this raises an important question about chaperone function in the nucleus. We performed a systematic analysis of the nuclear pool of heat-shock protein 90. Three orthogonal and independent analyses led us to the core functional interactome of HSP90. Computational and biochemical analyses identify host cell factor C1 (HCFC1) as a transcriptional regulator that depends on HSP90 for its stability. HSP90 was required to maintain the expression of HCFC1-targeted cell-cycle genes. The regulatory nexus between HSP90 and the HCFC1 module identified in this study sheds light on the relevance of chaperones in the transcription of cell-cycle genes. Our study also suggests a therapeutic avenue of combining chaperone and transcription inhibitors for cancer treatment.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, cdc , HSP90 Heat-Shock Proteins/metabolism , Host Cell Factor C1/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/genetics , Chromatin Immunoprecipitation Sequencing , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cytosol/metabolism , Databases, Genetic , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Host Cell Factor C1/genetics , Humans , Mice , Protein Binding , Protein Interaction Maps , RNA-Seq
9.
Proc Natl Acad Sci U S A ; 116(30): 14961-14970, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31296563

ABSTRACT

O-GlcNAc transferase (OGT) is an X-linked gene product that is essential for normal development of the vertebrate embryo. It catalyses the O-GlcNAc posttranslational modification of nucleocytoplasmic proteins and proteolytic maturation of the transcriptional coregulator Host cell factor 1 (HCF1). Recent studies have suggested that conservative missense mutations distal to the OGT catalytic domain lead to X-linked intellectual disability in boys, but it is not clear if this is through changes in the O-GlcNAc proteome, loss of protein-protein interactions, or misprocessing of HCF1. Here, we report an OGT catalytic domain missense mutation in monozygotic female twins (c. X:70779215 T > A, p. N567K) with intellectual disability that allows dissection of these effects. The patients show limited IQ with developmental delay and skewed X-inactivation. Molecular analyses revealed decreased OGT stability and disruption of the substrate binding site, resulting in loss of catalytic activity. Editing this mutation into the Drosophila genome results in global changes in the O-GlcNAc proteome, while in mouse embryonic stem cells it leads to loss of O-GlcNAcase and delayed differentiation down the neuronal lineage. These data imply that catalytic deficiency of OGT could contribute to X-linked intellectual disability.


Subject(s)
Catalytic Domain , Genetic Diseases, X-Linked/genetics , Intellectual Disability/genetics , Loss of Function Mutation , N-Acetylglucosaminyltransferases/genetics , Animals , Cell Line , Drosophila , Female , Genetic Diseases, X-Linked/pathology , Host Cell Factor C1/metabolism , Humans , Intellectual Disability/pathology , Mice , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Neurogenesis , Point Mutation , Twins, Monozygotic
10.
Cell Oncol (Dordr) ; 42(5): 579-589, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31183804

ABSTRACT

BACKGROUND: The p53 and HSF1 transcription factors are key players in cellular responses to stress. They activate important signaling pathways triggering adaptive mechanisms that maintain cellular homeostasis. HSF1 is mainly activated by proteotoxic stress, and its induction leads to the synthesis of chaperones that provide proteome integrity. The p53 protein, which is primarily activated in response to DNA damage, causes cell cycle arrest allowing for DNA repair or directs cells to apoptosis, thereby maintaining genome integrity. Both signaling pathways are also involved in neoplastic transformation and tumor progression. Loss of tumor suppressor abilities of the wild-type p53 protein results in oncogenesis, whereas proper HSF1 action, though non-oncogenic itself, actively supports this process. CONCLUSIONS: Here, we describe in detail the interplay between the p53 and HSF1 signaling pathways, with particular emphasis on the molecular mechanisms involved, as well as their importance for normal cellular behavior, cancer development, the effectiveness of anti-cancer therapies and their toxicity. Detailed knowledge of the complex interplay between HSF1 and p53 may form a basis for the design of new protocols for cancer treatment.


Subject(s)
Carcinogenesis/genetics , Host Cell Factor C1/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Carcinogenesis/metabolism , Cellular Senescence/genetics , Cytoprotection/genetics , Cytoprotection/physiology , DNA Repair , Disease Progression , Humans , Immediate-Early Proteins/metabolism , Neovascularization, Pathologic/metabolism , Nuclear Proteins/metabolism , Oncogene Addiction/genetics , Signal Transduction , Stress, Physiological/genetics , Tumor Suppressor Protein p53/genetics
11.
Theranostics ; 9(8): 2183-2197, 2019.
Article in English | MEDLINE | ID: mdl-31149037

ABSTRACT

O-GlcNAc transferase (OGT) is overexpressed in aggressive prostate cancer. OGT modifies intra-cellular proteins via single sugar conjugation (O-GlcNAcylation) to alter their activity. We recently discovered the first fast-acting OGT inhibitor OSMI-2. Here, we probe the stability and function of the chromatin O-GlcNAc and identify transcription factors that coordinate with OGT to promote proliferation of prostate cancer cells. Methods: Chromatin immunoprecipitation (ChIP) coupled to sequencing (seq), formaldehyde-assisted isolation of regulatory elements, RNA-seq and reverse-phase protein arrays (RPPA) were used to study the importance of OGT for chromatin structure and transcription. Mass spectrometry, western blot, RT-qPCR, cell cycle analysis and viability assays were used to establish the role of OGT for MYC-related processes. Prostate cancer patient data profiled for both mRNA and protein levels were used to validate findings. Results: We show for the first time that OGT inhibition leads to a rapid loss of O-GlcNAc chromatin mark. O-GlcNAc ChIP-seq regions overlap with super-enhancers (SE) and MYC binding sites. OGT inhibition leads to down-regulation of SE-dependent genes. We establish the first O-GlcNAc chromatin consensus motif, which we use as a bait for mass spectrometry. By combining the proteomic data from oligonucleotide enrichment with O-GlcNAc and MYC ChIP-mass spectrometry, we identify host cell factor 1 (HCF-1) as an interaction partner of MYC. Inhibition of OGT disrupts this interaction and compromises MYC's ability to confer androgen-independent proliferation to prostate cancer cells. We show that OGT is required for MYC-mediated stabilization of mitotic proteins, including Cyclin B1, and/or the increased translation of their coding transcripts. This implies that increased expression of mRNA is not always required to achieve increased protein expression and confer aggressive phenotype. Indeed, high expression of Cyclin B1 protein has strong predictive value in prostate cancer patients (p=0.000014) while mRNA does not. Conclusions: OGT promotes SE-dependent gene expression. OGT activity is required for the interaction between MYC and HCF-1 and expression of MYC-regulated mitotic proteins. These features render OGT essential for the androgen-independent, MYC-driven proliferation of prostate cancer cells. Androgen-independency is the major mechanism of prostate cancer progression, and our study identifies OGT as an essential mediator in this process.


Subject(s)
Cell Proliferation , N-Acetylglucosaminyltransferases/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Chromatin/metabolism , Chromatin Assembly and Disassembly , Cyclin B1/genetics , Cyclin B1/metabolism , Enhancer Elements, Genetic , Host Cell Factor C1/genetics , Host Cell Factor C1/metabolism , Humans , Male , Mice , N-Acetylglucosaminyltransferases/genetics , Prostatic Neoplasms/genetics , Transcriptional Activation
12.
Nucleic Acids Res ; 47(11): 5792-5808, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31049581

ABSTRACT

HCF-2 is a member of the host-cell-factor protein family, which arose in early vertebrate evolution as a result of gene duplication. Whereas its paralog, HCF-1, is known to act as a versatile chromatin-associated protein required for cell proliferation and differentiation, much less is known about HCF-2. Here, we show that HCF-2 is broadly present in human and mouse cells, and possesses activities distinct from HCF-1. Unlike HCF-1, which is excluded from nucleoli, HCF-2 is nucleolar-an activity conferred by one and a half C-terminal Fibronectin type 3 repeats and inhibited by the HCF-1 nuclear localization signal. Elevated HCF-2 synthesis in HEK-293 cells results in phenotypes reminiscent of HCF-1-depleted cells, including inhibition of cell proliferation and mitotic defects. Furthermore, increased HCF-2 levels in HEK-293 cells lead to inhibition of cell proliferation and metabolism gene-expression programs with parallel activation of differentiation and morphogenesis gene-expression programs. Thus, the HCF ancestor appears to have evolved into a small two-member protein family possessing contrasting nuclear versus nucleolar localization, and cell proliferation and differentiation functions.


Subject(s)
Gene Expression Profiling , Host Cell Factor C1/physiology , Transcription Factors/physiology , Animals , Cell Line , Cell Line, Tumor , Cell Nucleolus , Cell Proliferation , Chromatin/chemistry , Fibroblasts/metabolism , Gene Duplication , HEK293 Cells , HeLa Cells , Host Cell Factor C1/metabolism , Humans , Jurkat Cells , MCF-7 Cells , Mice , Mitosis , Nuclear Localization Signals/metabolism , Phenotype , Plasmids/metabolism , RNA, Small Interfering/metabolism , Transcription Factors/metabolism
13.
Cell ; 177(3): 722-736.e22, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30955890

ABSTRACT

Insulin receptor (IR) signaling is central to normal metabolic control and dysregulated in prevalent chronic diseases. IR binds insulin at the cell surface and transduces rapid signaling via cytoplasmic kinases. However, mechanisms mediating long-term effects of insulin remain unclear. Here, we show that IR associates with RNA polymerase II in the nucleus, with striking enrichment at promoters genome-wide. The target genes were highly enriched for insulin-related functions including lipid metabolism and protein synthesis and diseases including diabetes, neurodegeneration, and cancer. IR chromatin binding was increased by insulin and impaired in an insulin-resistant disease model. Promoter binding by IR was mediated by coregulator host cell factor-1 (HCF-1) and transcription factors, revealing an HCF-1-dependent pathway for gene regulation by insulin. These results show that IR interacts with transcriptional machinery at promoters and identify a pathway regulating genes linked to insulin's effects in physiology and disease.


Subject(s)
Gene Expression Regulation , Genome-Wide Association Study , Receptor, Insulin/metabolism , Animals , Cell Line, Tumor , Chromatin/metabolism , Gene Expression Regulation/drug effects , Host Cell Factor C1/antagonists & inhibitors , Host Cell Factor C1/genetics , Host Cell Factor C1/metabolism , Humans , Insulin/metabolism , Insulin/pharmacology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Protein Binding , Protein Subunits/metabolism , RNA Interference , RNA Polymerase II/metabolism , RNA, Small Interfering/metabolism , Receptor, Insulin/chemistry , Signal Transduction/drug effects
14.
Cell Death Dis ; 9(11): 1118, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389914

ABSTRACT

Although additional sex combs-like 1 (ASXL1) has been extensively described in hematologic malignancies, little is known about the molecular role of ASXL1 in organ development. Here, we show that Asxl1 ablation in mice results in postnatal lethality due to cyanosis, a respiratory failure. This lung defect is likely caused by higher proliferative potential and reduced expression of surfactant proteins, leading to reduced air space and defective lung maturation. By microarray analysis, we identified E2F1-responsive genes, including Nmyc, as targets repressed by Asxl1. Nmyc and Asxl1 are reciprocally expressed during the fetal development of normal mouse lungs, whereas Nmyc downregulation is impaired in Asxl1-deficient lungs. Together with E2F1 and ASXL1, host cell factor 1 (HCF-1), purified as an Asxl1-bound protein, is recruited to the E2F1-binding site of the Nmyc promoter. The interaction occurs between the C-terminal region of Asxl1 and the N-terminal Kelch domain of HCF-1. Trimethylation (me3) of histone H3 lysine 27 (H3K27) is enriched in the Nmyc promoter upon Asxl1 overexpression, whereas it is downregulated in Asxl1-deleted lung and -depleted A549 cells, similar to H3K9me3, another repressive histone marker. Overall, these findings suggest that Asxl1 modulates proliferation of lung epithelial cells via the epigenetic repression of Nmyc expression, deficiency of which may cause hyperplasia, leading to dyspnea.


Subject(s)
E2F1 Transcription Factor/genetics , Epigenetic Repression , Epithelial Cells/metabolism , Lung/metabolism , N-Myc Proto-Oncogene Protein/genetics , Repressor Proteins/genetics , Respiratory Insufficiency/genetics , A549 Cells , Animals , E2F1 Transcription Factor/metabolism , Embryo, Mammalian , Epithelial Cells/pathology , Fetus , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, Lethal , HEK293 Cells , Histones/genetics , Histones/metabolism , Host Cell Factor C1/genetics , Host Cell Factor C1/metabolism , Humans , Lung/growth & development , Lung/pathology , Mice , Mice, Knockout , N-Myc Proto-Oncogene Protein/metabolism , Organogenesis/genetics , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/deficiency , Respiratory Insufficiency/metabolism , Respiratory Insufficiency/pathology , Signal Transduction
15.
J Biol Chem ; 293(46): 17754-17768, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30224358

ABSTRACT

O-Linked GlcNAc transferase (OGT) possesses dual glycosyltransferase-protease activities. OGT thereby stably glycosylates serines and threonines of numerous proteins and, via a transient glutamate glycosylation, cleaves a single known substrate-the so-called HCF-1PRO repeat of the transcriptional co-regulator host-cell factor 1 (HCF-1). Here, we probed the relationship between these distinct glycosylation and proteolytic activities. For proteolysis, the HCF-1PRO repeat possesses an important extended threonine-rich region that is tightly bound by the OGT tetratricopeptide-repeat (TPR) region. We report that linkage of this HCF-1PRO-repeat, threonine-rich region to heterologous substrate sequences also potentiates robust serine glycosylation with the otherwise poor Rp-αS-UDP-GlcNAc diastereomer phosphorothioate and UDP-5S-GlcNAc OGT co-substrates. Furthermore, it potentiated proteolysis of a non-HCF-1PRO-repeat cleavage sequence, provided it contained an appropriately positioned glutamate residue. Using serine- or glutamate-containing HCF-1PRO-repeat sequences, we show that proposed OGT-based or UDP-GlcNAc-based serine-acceptor residue activation mechanisms can be circumvented independently, but not when disrupted together. In contrast, disruption of both proposed activation mechanisms even in combination did not inhibit OGT-mediated proteolysis. These results reveal a multiplicity of OGT glycosylation strategies, some leading to proteolysis, which could be targets of alternative molecular regulatory strategies.


Subject(s)
Endopeptidases/metabolism , Host Cell Factor C1/metabolism , N-Acetylglucosaminyltransferases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Endopeptidases/genetics , Glycosylation , Host Cell Factor C1/genetics , Humans , Molecular Dynamics Simulation , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Mutation , N-Acetylglucosaminyltransferases/genetics , Proteolysis , Stereoisomerism , Substrate Specificity , Uridine Diphosphate N-Acetylglucosamine/analogs & derivatives , Uridine Diphosphate N-Acetylglucosamine/metabolism
16.
Mol Cell Biol ; 38(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29941490

ABSTRACT

Cancer cells often heavily depend on the ubiquitin-proteasome system (UPS) for their growth and survival. Irrespective of their strong dependence on the proteasome activity, cancer cells, except for multiple myeloma, are mostly resistant to proteasome inhibitors. A major cause of this resistance is the proteasome bounce-back response mediated by NRF1, a transcription factor that coordinately activates proteasome subunit genes. To identify new targets for efficient suppression of UPS, we explored, using immunoprecipitation and mass spectrometry, the possible existence of nuclear proteins that cooperate with NRF1 and identified O-linked N-acetylglucosamine transferase (OGT) and host cell factor C1 (HCF-1) as two proteins capable of forming a complex with NRF1. O-GlcNAcylation catalyzed by OGT was essential for NRF1 stabilization and consequent upregulation of proteasome subunit genes. Meta-analysis of breast and colorectal cancers revealed positive correlations in the relative protein abundance of OGT and proteasome subunits. OGT inhibition was effective at sensitizing cancer cells to a proteasome inhibitor both in culture cells and a xenograft mouse model. Since active O-GlcNAcylation is a feature of cancer metabolism, our study has clarified a novel linkage between cancer metabolism and UPS function and added a new regulatory axis to the regulation of the proteasome activity.


Subject(s)
NF-E2-Related Factor 1/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Proteasome Inhibitors/pharmacology , Acetylglucosamine/metabolism , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/physiology , Female , Glycosylation , HEK293 Cells , HeLa Cells , Host Cell Factor C1/chemistry , Host Cell Factor C1/genetics , Host Cell Factor C1/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , NF-E2-Related Factor 1/chemistry , NF-E2-Related Factor 1/genetics , Neoplasms/genetics , Nuclear Respiratory Factor 1 , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Interaction Domains and Motifs , Protein Stability , Ubiquitin/metabolism , Xenograft Model Antitumor Assays , beta-Transducin Repeat-Containing Proteins/chemistry , beta-Transducin Repeat-Containing Proteins/metabolism
17.
J Virol ; 92(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29899098

ABSTRACT

Following productive infection, bovine herpesvirus 1 (BoHV-1) establishes latency in sensory neurons. As in other alphaherpesviruses, expression of BoHV-1 immediate early (IE) genes is regulated by an enhancer complex containing the viral IE activator VP16, the cellular transcription factor Oct-1, and transcriptional coactivator HCF-1, which is assembled on an IE enhancer core element (TAATGARAT). Expression of the IE transcription unit that encodes the viral IE activators bICP0 and bICP4 may also be induced by the activated glucocorticoid receptor (GR) via two glucocorticoid response elements (GREs) located upstream of the enhancer core. Strikingly, lytic infection and reactivation from latency are consistently enhanced by glucocorticoid treatment in vivo As the coactivator HCF-1 is essential for IE gene expression of alphaherpesviruses and recruited by multiple transcription factors, we tested whether HCF-1 is required for glucocorticoid-induced IE gene expression. Depletion of HCF-1 reduced GR-mediated activation of the IE promoter in mouse neuroblastoma cells (Neuro-2A). More importantly, HCF-1-mediated GR activation of the promoter was dependent on the presence of GRE sites but independent of the TAATGARAT enhancer core element. HCF-1 was also recruited to the GRE region of a promoter lacking the enhancer core, consistent with a direct role of the coactivator in mediating GR-induced transcription. Similarly, during productive lytic infection, HCF-1 and GR occupied the IE region containing the GREs. These studies indicate HCF-1 is critical for GR activation of the viral IE genes and suggests that glucocorticoid induction of viral reactivation proceeds via an HCF-1-GR mechanism in the absence of the viral IE activator VP16.IMPORTANCE BoHV-1 transcription is rapidly activated during stress-induced reactivation from latency. The immediate early transcription unit 1 (IEtu1) promoter is regulated by the GR via two GREs. The IEtu1 promoter regulates expression of two viral transcriptional regulatory proteins, infected cell proteins 0 and 4 (bICP0 and bICP4), and thus must be stimulated during reactivation. This study demonstrates that activation of the IEtu1 promoter by the synthetic corticosteroid dexamethasone requires HCF-1. Interestingly, the GRE sites, but not the IE enhancer core element (TAATGARAT), were required for HCF-1-mediated GR promoter activation. The GR and HCF-1 were recruited to the IEtu1 promoter in transfected and infected cells. Collectively, these studies indicate that HCF-1 is critical for GR activation of the viral IE genes and suggest that an HCF-1-GR complex can stimulate the IEtu1 promoter in the absence of the viral IE activator VP16.


Subject(s)
Gene Expression Regulation, Viral , Genes, Immediate-Early , Glucocorticoids/metabolism , Herpesvirus 1, Bovine/physiology , Host Cell Factor C1/metabolism , Receptors, Glucocorticoid/metabolism , Transcription, Genetic , Animals , Cell Line , Gene Knockdown Techniques , Host Cell Factor C1/genetics , Mice , Neurons/virology
18.
Cell Rep ; 21(6): 1562-1573, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29117561

ABSTRACT

Ronin (THAP11), a DNA-binding protein that evolved from a primordial DNA transposon by molecular domestication, recognizes a hyperconserved promoter sequence to control developmentally and metabolically essential genes in pluripotent stem cells. However, it remains unclear whether Ronin or related THAP proteins perform similar functions in development. Here, we present evidence that Ronin functions within the nascent heart as it arises from the mesoderm and forms a four-chambered organ. We show that Ronin is vital for cardiogenesis during midgestation by controlling a set of critical genes. The activity of Ronin coincided with the recruitment of its cofactor, Hcf-1, and the elevation of H3K4me3 levels at specific target genes, suggesting the involvement of an epigenetic mechanism. On the strength of these findings, we propose that Ronin activity during cardiogenesis offers a template to understand how important gene programs are sustained across different cell types within a developing organ such as the heart.


Subject(s)
Heart/growth & development , Repressor Proteins/metabolism , Animals , Bradycardia/metabolism , Bradycardia/physiopathology , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Chromatin Immunoprecipitation , Echocardiography , Embryo, Mammalian/metabolism , Embryonic Development , Gene Expression Regulation, Developmental , Heart/diagnostic imaging , Histones/genetics , Histones/metabolism , Homeobox Protein Nkx-2.5/deficiency , Homeobox Protein Nkx-2.5/genetics , Host Cell Factor C1/genetics , Host Cell Factor C1/metabolism , Methylation , Mice , Mice, Knockout , Microscopy, Fluorescence , Myosin Heavy Chains/deficiency , Myosin Heavy Chains/genetics , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/genetics , Transcription Initiation Site
19.
Free Radic Biol Med ; 110: 196-205, 2017 09.
Article in English | MEDLINE | ID: mdl-28625484

ABSTRACT

The Nrf1 (Nuclear factor E2-related factor 1) transcription factor performs a critical role in regulating cellular homeostasis. Using a proteomic approach, we identified Host Cell Factor-1 (HCF1), a co-regulator of transcription, and O-GlcNAc transferase (OGT), the enzyme that mediates protein O-GlcNAcylation, as cellular partners of Nrf1a, an isoform of Nrf1. Nrf1a directly interacts with HCF1 through the HCF1 binding motif (HBM), while interaction with OGT is mediated through HCF1. Overexpression of HCF1 and OGT leads to increased Nrf1a protein stability. Addition of O-GlcNAc decreases ubiquitination and degradation of Nrf1a. Transcriptional activation by Nrf1a is increased by OGT overexpression and treatment with PUGNAc. Together, these data suggest that OGT can act as a regulator of Nrf1a.


Subject(s)
Host Cell Factor C1/metabolism , N-Acetylglucosaminyltransferases/metabolism , Nuclear Respiratory Factor 1/metabolism , Protein Processing, Post-Translational , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/pharmacology , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Gene Expression , Glycosylation , HEK293 Cells , Host Cell Factor C1/chemistry , Host Cell Factor C1/genetics , Humans , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/genetics , Nuclear Respiratory Factor 1/chemistry , Nuclear Respiratory Factor 1/genetics , Oximes/pharmacology , Phenylcarbamates/pharmacology , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcriptional Activation/drug effects , Transfection , Ubiquitination
20.
Hum Mol Genet ; 26(15): 2975-2983, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28486698

ABSTRACT

Thanatos-associated protein domain containing, apoptosis-associated protein 1 (THAP1), the gene mutated in DYT6 dystonia, encodes a transcription factor. While the N-terminal THAP domain allows for specific DNA-binding, the functional relevance of the other regions is largely unknown. The C-terminus contains a 4-amino-acid-spanning host cell factor 1 (HCFC1)-binding domain (HBM) that mediates the interaction with HCFC1. Interestingly, three mutations affecting the HBM (p.N136S, p.N136K, p.Y137C) have been reported in dystonia patients. We investigated the consequences of these mutations on the interaction of THAP1 with HCFC1 and demonstrated that all three mutations abolished HCFC1-THAP1 complex formation. Notably, HCFC1 co-localization was found in >90% of the almost 3,500 chromatin regions loaded with THAP1 in publicly available genome-wide ChIP data. By siRNA-mediated depletion of HCFC1, we detected an increase of THAP1 expression, indicating a co-repressor activity of HCFC1 for THAP1. Quantitative ChIP on selected promoters revealed that none of the mutations significantly decreased the DNA-binding ability of THAP1 while HCFC1 binding was highly reduced. Our findings indicate a THAP1-mediated recruitment of HCFC1 to THAP1 target sites. Of note, dystonia-causing mutations within the HBM in THAP1 abolished this interaction. Thus, we demonstrate disrupted THAP1-HCFC1 complex formation as another mechanism of dystonia-causing mutations leading to transcriptional dysregulation.


Subject(s)
Apoptosis Regulatory Proteins/genetics , DNA-Binding Proteins/genetics , Dystonia/genetics , Host Cell Factor C1/genetics , Nuclear Proteins/genetics , Amino Acid Motifs , Apoptosis Regulatory Proteins/metabolism , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , DNA-Binding Proteins/metabolism , Dystonic Disorders/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Host Cell Factor C1/metabolism , Host Factor 1 Protein , Humans , Mutation/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...