Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Article in English | MEDLINE | ID: mdl-39073416

ABSTRACT

Glioblastoma (GBM) is the most aggressive form of brain cancer, characterized by rapid growth and invasion into surrounding brain tissue. Ubiquitin-specific protease 9X (USP9X) has emerged as a key regulator in various cancers, but its role in GBM pathogenesis remains unclear. Understanding the molecular mechanisms underlying USP9X modulation of GBM progression could unveil potential therapeutic targets for this deadly disease. The mRNA and protein levels were determined in GBM tissues and/or cells using quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting assays, respectively. Cell migration was evaluated through wound-healing assay, while cell proliferation was measured using colony formation and CCK-8 assays. Flow cytometry analysis was performed to quantify the CD206-positive macrophages to assess M2 polarization. Co-immunoprecipitation (Co-IP) assays were conducted to elucidate the association between USP9X and transformation/transcription domain-associated protein (TRRAP). Cycloheximide (CHX) treatment was used to determine the impact of USP9X on TRRAP protein stabilization. Furthermore, the effect of USP9X depletion on GBM cell malignancy was validated using a xenograft mouse model. We found that USP9X expression was elevated in GBM tissues and cells. Depletion of USP9X suppressed GBM cell migration, proliferation, and M2 macrophage polarization. Mechanistically, USP9X stabilized TRRAP through the deubiquitination pathway in GBM cells, and TRRAP mitigated the effects of USP9X silencing on GBM cell malignant phenotypes and M2 macrophage polarization. Moreover, silencing of USP9X inhibited tumor formation in vivo. Together, USP9X deubiquitinated TRRAP, thereby promoting glioblastoma cell proliferation, migration, and M2 macrophage polarization. These results highlight the potential of targeting the USP9X-TRRAP axis as a therapeutic strategy for GBM.

2.
Int J Mol Sci ; 24(9)2023 May 05.
Article in English | MEDLINE | ID: mdl-37175973

ABSTRACT

The heterotrimeric Tel2-Tti1-Tti2 or TTT complex is essential for cell viability and highly observed in eukaryotes. As the co-chaperone of ATR, ATM, DNA-PKcs, mTOR, SMG1, and TRRAP, the phosphatidylinositol 3-kinase-related kinases (PIKKs) and a group of large proteins of 300-500 kDa, the TTT plays crucial roles in genome stability, cell proliferation, telomere maintenance, and aging. Most of the protein kinases in the kinome are targeted by co-chaperone Cdc37 for proper folding and stability. Like Cdc37, accumulating evidence has established the mechanism by which the TTT interacts with chaperone Hsp90 via R2TP (Rvb1-Rvb2-Tah1-Pih1) complex or other proteins for co-translational maturation of the PIKKs. Recent structural studies have revealed the α-solenoid structure of the TTT and its interactions with the R2TP complex, which shed new light on the co-chaperone mechanism and provide new research opportunities. A series of mutations of the TTT have been identified that cause disease syndrome with neurodevelopmental defects, and misregulation of the TTT has been shown to contribute to myeloma, colorectal, and non-small-cell lung cancers. Surprisingly, Tel2 in the TTT complex has recently been found to be a target of ivermectin, an antiparasitic drug that has been used by millions of patients. This discovery provides mechanistic insight into the anti-cancer effect of ivermectin and thus promotes the repurposing of this Nobel-prize-winning medicine for cancer chemotherapy. Here, we briefly review the discovery of the TTT complex, discuss the recent studies, and describe the perspectives for future investigation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , HSP90 Heat-Shock Proteins/metabolism , Ivermectin , Molecular Chaperones/metabolism , Telomere-Binding Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism
3.
Int J Mol Sci ; 24(7)2023 Mar 26.
Article in English | MEDLINE | ID: mdl-37047234

ABSTRACT

NANOG, a stemness-associated transcription factor, is highly expressed in many cancers and plays a critical role in regulating tumorigenicity. Transformation/transcription domain-associated protein (TRRAP) has been reported to stimulate the tumorigenic potential of cancer cells and induce the gene transcription of NANOG. This study aimed to investigate the role of the TRRAP-NANOG signaling pathway in the tumorigenicity of cancer stem cells. We found that TRRAP overexpression specifically increases NANOG protein stability by interfering with NANOG ubiquitination mediated by FBXW8, an E3 ubiquitin ligase. Mapping of NANOG-binding sites using deletion mutants of TRRAP revealed that a domain of TRRAP (amino acids 1898-2400) is responsible for binding to NANOG and that the overexpression of this TRRAP domain abrogated the FBXW8-mediated ubiquitination of NANOG. TRRAP knockdown decreased the expression of CD44, a cancer stem cell marker, and increased the expression of P53, a tumor suppressor gene, in HCT-15 colon cancer cells. TRRAP depletion attenuated spheroid-forming ability and cisplatin resistance in HCT-15 cells, which could be rescued by NANOG overexpression. Furthermore, TRRAP knockdown significantly reduced tumor growth in a murine xenograft transplantation model, which could be reversed by NANOG overexpression. Together, these results suggest that TRRAP plays a pivotal role in the regulation of the tumorigenic potential of colon cancer cells by modulating NANOG protein stability.


Subject(s)
Colonic Neoplasms , Animals , Humans , Mice , Carcinogenesis/genetics , Cell Line, Tumor , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/metabolism , Protein Stability
4.
Comput Struct Biotechnol J ; 21: 472-484, 2023.
Article in English | MEDLINE | ID: mdl-36618986

ABSTRACT

The adult hippocampal neurogenesis plays a vital role in the function of the central nervous system (CNS), including memory consolidation, cognitive flexibility, emotional function, and social behavior. The deficiency of adult neural stem cells (aNSCs) in maintaining the quiescence and entering cell cycle, self-renewal and differentiation capacity is detrimental to the functional integrity of neurons and cognition of the adult brain. Histone acetyltransferase (HAT) and histone deacetylase (HDAC) have been shown to modulate brain functionality and are important for embryonic neurogenesis via regulation of gene transcription. We showed previously that Trrap, an adapter for several HAT complexes, is required for Sp1 transcriptional control of the microtubule dynamics in neuronal cells. Here, we find that Trrap deletion compromises self-renewal and differentiation of aNSCs in mice and in cultures. We find that the acetylation status of lysine residues K16, K19, K703 and K639 all fail to overcome Trrap-deficiency-incurred instability of Sp1, indicating a scaffold role of Trrap. Interestingly, the deacetylation of Sp1 at K639 and K703 greatly increases Sp1 binding to the promoter of target genes, which antagonizes Trrap binding, and thereby elevates Sp1 activity. However, only deacetylated K639 is refractory to Trrap deficiency and corrects the differentiation defects of Trrap-deleted aNSCs. We demonstrate that the acetylation pattern at K639 by HATs dictates the role of Sp1 in the regulation of adult neurogenesis.

5.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830324

ABSTRACT

The members of the phosphatidylinositol 3-kinase-related kinase (PIKK) family play vital roles in multiple biological processes, including DNA damage response, metabolism, cell growth, mRNA decay, and transcription. TRRAP, as the only member lacking the enzymatic activity in this family, is an adaptor protein for several histone acetyltransferase (HAT) complexes and a scaffold protein for multiple transcription factors. TRRAP has been demonstrated to regulate various cellular functions in cell cycle progression, cell stemness maintenance and differentiation, as well as neural homeostasis. TRRAP is known to be an important orchestrator of many molecular machineries in gene transcription by modulating the activity of some key transcription factors, including E2F1, c-Myc, p53, and recently, Sp1. This review summarizes the biological and biochemical studies on the action mode of TRRAP together with the transcription factors, focusing on how TRRAP-HAT mediates the transactivation of Sp1-governing biological processes, including neurodegeneration.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Histone Acetyltransferases/genetics , Neurodegenerative Diseases/genetics , Neurogenesis/genetics , Nuclear Proteins/genetics , Sp1 Transcription Factor/genetics , Transcription, Genetic , Adaptor Proteins, Signal Transducing/metabolism , Animals , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Gene Expression Regulation , Histone Acetyltransferases/metabolism , Humans , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Nuclear Proteins/metabolism , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Sp1 Transcription Factor/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Elife ; 102021 02 17.
Article in English | MEDLINE | ID: mdl-33594975

ABSTRACT

Brain homeostasis is regulated by the viability and functionality of neurons. HAT (histone acetyltransferase) and HDAC (histone deacetylase) inhibitors have been applied to treat neurological deficits in humans; yet, the epigenetic regulation in neurodegeneration remains elusive. Mutations of HAT cofactor TRRAP (transformation/transcription domain-associated protein) cause human neuropathies, including psychosis, intellectual disability, autism, and epilepsy, with unknown mechanism. Here we show that Trrap deletion in Purkinje neurons results in neurodegeneration of old mice. Integrated transcriptomics, epigenomics, and proteomics reveal that TRRAP via SP1 conducts a conserved transcriptomic program. TRRAP is required for SP1 binding at the promoter proximity of target genes, especially microtubule dynamics. The ectopic expression of Stathmin3/4 ameliorates defects of TRRAP-deficient neurons, indicating that the microtubule dynamics is particularly vulnerable to the action of SP1 activity. This study unravels a network linking three well-known, but up-to-date unconnected, signaling pathways, namely TRRAP, HAT, and SP1 with microtubule dynamics, in neuroprotection.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Aging , Animals , Epigenesis, Genetic , Gene Deletion , Gene Expression Regulation , Mice , Mice, Mutant Strains , Microtubules/metabolism , Purkinje Cells/pathology , Signal Transduction
7.
Semin Cell Dev Biol ; 110: 51-60, 2021 02.
Article in English | MEDLINE | ID: mdl-32362381

ABSTRACT

Multiciliated cells (MCC) project dozens to hundreds of motile cilia from the cell surface to generate fluid flow across epithelial surfaces or turbulence to promote the transport of gametes. The MCC differentiation program is initiated by GEMC1 and MCIDAS, members of the geminin family, that activate key transcription factors, including p73 and FOXJ1, to control the multiciliogenesis program. To support the generation of multiple motile cilia, MCCs must undergo massive centriole amplification to generate a sufficient number of basal bodies (modified centrioles). This transcriptional program involves the generation of deuterosomes, unique structures that act as platforms to regulate centriole amplification, the reactivation of cell cycle programs to control centriole amplification and release, and extensive remodeling of the cytoskeleton. This review will focus on providing an overview of the transcriptional regulation of MCCs and its connection to key processes, in addition to highlighting exciting recent developments and open questions in the field.


Subject(s)
Cell Cycle Proteins/genetics , Centrioles/metabolism , Cilia/metabolism , Ciliopathies/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Cell Cycle/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Centrioles/ultrastructure , Cilia/ultrastructure , Ciliopathies/metabolism , Ciliopathies/pathology , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Humans , Signal Transduction , Transcription Factors/metabolism , Tumor Protein p73/genetics , Tumor Protein p73/metabolism
8.
Cancer Biol Med ; 17(3): 782-794, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32944406

ABSTRACT

Objective: Osteosarcoma is the most common primary malignant bone tumor. However, the survival of patients with osteosarcoma has remained unchanged during the past 30 years, owing to a lack of efficient therapeutic targets. Methods: We constructed a kinome-targeting CRISPR-Cas9 library containing 507 kinases and 100 nontargeting controls and screened the potential kinase targets in osteosarcoma. The CRISPR screening sequencing data were analyzed with the Model-based Analysis of Genome-wide CRISPR/Cas9 Knockout (MAGeCK) Python package. The functional data were applied in the 143B cell line through lenti-CRISPR-mediated gene knockout. The clinical significance of kinases in the survival of patients with osteosarcoma was analyzed in the R2: Genomics Analysis and Visualization Platform. Results: We identified 53 potential kinase targets in osteosarcoma. Among these targets, we analyzed 3 kinases, TRRAP, PKMYT1, and TP53RK, to validate their oncogenic functions in osteosarcoma. PKMYT1 and TP53RK showed higher expression in osteosarcoma than in normal bone tissue, whereas TRRAP showed no significant difference. High expression of all 3 kinases was associated with relatively poor prognosis in patients with osteosarcoma. Conclusions: Our results not only offer potential therapeutic kinase targets in osteosarcoma but also provide a paradigm for functional genetic screening by using a CRISPR-Cas9 library, including target design, library construction, screening workflow, data analysis, and functional validation. This method may also be useful in potentially accelerating drug discovery for other cancer types.


Subject(s)
Bone Neoplasms/metabolism , CRISPR-Cas Systems/genetics , Osteosarcoma/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Gene Knockout Techniques , Gene Library , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteosarcoma/genetics , Osteosarcoma/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism
9.
Clin Genet ; 96(4): 300-308, 2019 10.
Article in English | MEDLINE | ID: mdl-31231791

ABSTRACT

Hereditary non-syndromic hearing loss is the most common inherited sensory defect in humans. More than 40 genes have been identified as causative genes for autosomal dominant non-syndromic hearing loss (ADNSHL), but there are many other candidate genes that remain to be discovered. We aimed to identify the causative gene mutation for post-lingual progressive ADNSHL in a Chinese family. Whole-exome sequencing, bioinformatic analysis, and Sanger sequencing were used to verify the co-segregation of a novel pathogenic variant (NM_ 001244580, c.511C>T, p.Arg171Cys) in the TRansformation/tRanscription domain-Associated Protein gene associated with hearing loss in a three-generation Chinese family with ADNSHL). Additionally, three more novel variants of transformation/transcription domain associated protein (TRRAP) were detected in 66 sporadic cases of hearing loss. Morpholino oligonucleotides knockdown and clustered regularly interspaced short palindromic repeats/Cas9 knockout zebrafish were constructed to validate the genetic findings. Knockdown or knockout of TRRAP resulted in significant defects in the inner ear of zebrafish, indicating that TRRAP plays an important role in inner ear development. In conclusion, TRRAP (NM_ 001244580, c.511C>T, p.Arg171Cys) co-segregated with hearing loss in a Chinese family with ADNSHL, and TRRAP deficiency caused hearing disability in zebrafish, suggesting TRRAP is a gene associated with ADNSHL.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Genes, Dominant , Genetic Association Studies , Genetic Predisposition to Disease , Hearing Loss/diagnosis , Hearing Loss/genetics , Mutation , Nuclear Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Female , Gene Targeting , Genetic Association Studies/methods , Genotype , Humans , Male , Nuclear Proteins/metabolism , Pedigree , Phenotype , Sequence Analysis, DNA , Tomography, X-Ray Computed , Exome Sequencing , Zebrafish
10.
Am J Hum Genet ; 104(3): 530-541, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30827496

ABSTRACT

Acetylation of the lysine residues in histones and other DNA-binding proteins plays a major role in regulation of eukaryotic gene expression. This process is controlled by histone acetyltransferases (HATs/KATs) found in multiprotein complexes that are recruited to chromatin by the scaffolding subunit transformation/transcription domain-associated protein (TRRAP). TRRAP is evolutionarily conserved and is among the top five genes intolerant to missense variation. Through an international collaboration, 17 distinct de novo or apparently de novo variants were identified in TRRAP in 24 individuals. A strong genotype-phenotype correlation was observed with two distinct clinical spectra. The first is a complex, multi-systemic syndrome associated with various malformations of the brain, heart, kidneys, and genitourinary system and characterized by a wide range of intellectual functioning; a number of affected individuals have intellectual disability (ID) and markedly impaired basic life functions. Individuals with this phenotype had missense variants clustering around the c.3127G>A p.(Ala1043Thr) variant identified in five individuals. The second spectrum manifested with autism spectrum disorder (ASD) and/or ID and epilepsy. Facial dysmorphism was seen in both groups and included upslanted palpebral fissures, epicanthus, telecanthus, a wide nasal bridge and ridge, a broad and smooth philtrum, and a thin upper lip. RNA sequencing analysis of skin fibroblasts derived from affected individuals skin fibroblasts showed significant changes in the expression of several genes implicated in neuronal function and ion transport. Thus, we describe here the clinical spectrum associated with TRRAP pathogenic missense variants, and we suggest a genotype-phenotype correlation useful for clinical evaluation of the pathogenicity of the variants.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autistic Disorder/etiology , Intellectual Disability/etiology , Mutation, Missense , Nuclear Proteins/genetics , Adolescent , Adult , Amino Acid Sequence , Autistic Disorder/metabolism , Autistic Disorder/pathology , Child , Child, Preschool , Female , Genetic Association Studies , Humans , Infant , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Prognosis , Sequence Homology , Syndrome , Young Adult
11.
Mol Cell ; 72(5): 836-848.e7, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30415952

ABSTRACT

Transforming members of the MYC family (MYC, MYCL1, and MYCN) encode transcription factors containing six highly conserved regions, termed MYC homology boxes (MBs). By conducting proteomic profiling of the MB interactomes, we demonstrate that half of the MYC interactors require one or more MBs for binding. Comprehensive phenotypic analyses reveal that two MBs, MB0 and MBII, are universally required for transformation. MBII mediates interactions with acetyltransferase-containing complexes, enabling histone acetylation, and is essential for MYC-dependent tumor initiation. By contrast, MB0 mediates interactions with transcription elongation factors via direct binding to the general transcription factor TFIIF. MB0 is dispensable for tumor initiation but is a major accelerator of tumor growth. Notably, the full transforming activity of MYC can be restored by co-expression of the non-transforming MB0 and MBII deletion proteins, indicating that these two regions confer separate molecular functions, both of which are required for oncogenic MYC activity.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors, TFII/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Profiling , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Protein Binding , Protein Domains , Protein Interaction Mapping , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Survival Analysis , Transcription Factors, TFII/metabolism , Tumor Burden , Xenograft Model Antitumor Assays
12.
Virology ; 512: 172-179, 2017 12.
Article in English | MEDLINE | ID: mdl-28965007

ABSTRACT

Cellular transformation by adenovirus E1A requires targeting TRRAP, a scaffold protein which helps assemble histone acetyltransferase complexes, including the NuA4 complex. We recently reported that E1A and E1A 1-80 (N-terminal 80 aa) promote association of the proto-oncogene product MYC with the NuA4 complex. The E1A N-terminal TRRAP-targeting (ET) domain is required for E1A 1-80 to interact with the NuA4 complex. We demonstrate that an ET-MYC fusion associates with the NuA4 complex more efficiently than does MYC alone. Because MYC regulates genes for multiple cellular pathways, we performed global RNA-sequence analysis of cells expressing MYC or ET-MYC, and identified a panel of genes (262) preferentially activated by ET-MYC and significantly enriched in genes involved in gene expression and ribosome biogenesis, suggesting that E1A enhances MYC association with the NuA4 complex to activate a set of MYC target genes likely involved in cellular proliferation and cellular transformation by E1A and by MYC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenovirus E1A Proteins/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Ribosomes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenovirus E1A Proteins/genetics , Cell Line , Gene Expression Regulation/physiology , Humans , Multigene Family , Multiprotein Complexes/genetics , Nuclear Proteins/genetics , Protein Binding , Protein Interaction Domains and Motifs , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics
13.
Development ; 144(4): 612-623, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28196804

ABSTRACT

Adult stem cells uphold a delicate balance between quiescent and active states, a deregulation of which can lead to age-associated diseases such as cancer. In Drosophila, intestinal stem cell (ISC) proliferation is tightly regulated and mis-regulation is detrimental to intestinal homeostasis. Various factors are known to govern ISC behavior; however, transcriptional changes in ISCs during aging are still unclear. RNA sequencing of young and old ISCs newly identified Nipped-A, a subunit of histone acetyltransferase complexes, as a regulator of ISC proliferation that is upregulated in old ISCs. We show that Nipped-A is required for maintaining the proliferative capacity of ISCs during aging and in response to tissue-damaging or tumorigenic stimuli. Interestingly, Drosophila Myc cannot compensate for the effect of the loss of Nipped-A on ISC proliferation. Nipped-A seems to be a superordinate regulator of ISC proliferation, possibly by coordinating different processes including modifying the chromatin landscape of ISCs and progenitors.


Subject(s)
Adult Stem Cells/cytology , Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Gene Expression Regulation, Developmental , Intestines/cytology , Transcription Factors/physiology , Aging , Animals , Cell Cycle , Cell Differentiation , Cell Proliferation , Cell Separation , Chromatin/metabolism , Flow Cytometry , Green Fluorescent Proteins/metabolism , Histones/metabolism , Homeostasis , Phenotype , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Sequence Analysis, RNA , Signal Transduction
14.
Genes Cancer ; 8(11-12): 752-761, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29321817

ABSTRACT

The proto-oncogene MYC is a transcription factor over-expressed in many cancers and required for cell survival. Its function is regulated by histone acetyltransferase (HAT) complexes, such as the GCN5 complex and the NuA4/Tip60 complex. However, the roles of the HAT complexes during MYC function in cancer have not been well characterized. We recently showed that adenovirus E1A and its N-terminal 80 aa region, E1A 1-80, interact with the NuA4 complex, through the E1A TRRAP-targeting (ET) domain, and enhance MYC association with the NuA4 complex. We show here that the ET domain mainly targets the MYC-NuA4 complex. By global gene expression analysis using E1A 1-80 and deletion mutants, we have identified a panel of genes activated by targeting the MYC-NuA4 complex and notably enriched for genes involved in ribosome biogenesis and gene expression. A second panel of genes is activated by E1A 1-80 targeting of both the MYC-NuA4 complex and p300, and is enriched for genes involved in DNA replication and cell cycle processes. Both panels of genes are highly expressed in cancer cells. Since the ET domain is essential for E1A-mediated cellular transformation, our results suggest that MYC and the NuA4 complex function cooperatively in cell transformation and cancer.

15.
Virology ; 499: 178-184, 2016 12.
Article in English | MEDLINE | ID: mdl-27664947

ABSTRACT

The adenovirus E1A 243R oncoprotein targets TRRAP, a scaffold protein that assembles histone acetyltransferase (HAT) complexes, such as the NuA4/Tip60 complex which mediates transcriptional activity of the proto-oncogene MYC and helps determine the cancer cell phenotype. How E1A transforms cells through TRRAP remains obscure. We performed proteomic analysis with the N-terminal transcriptional repression domain of E1A 243R (E1A 1-80) and showed that E1A 1-80 interacts with TRRAP, p400, and three other members of the NuA4 complex - DMAP1, RUVBL1 and RUVBL2 - not previously shown to associate with E1A 243R. E1A 1-80 interacts with these NuA4 components and MYC through the E1A TRRAP-targeting domain. E1A 243R association with the NuA4 complex was demonstrated by co-immunoprecipitation and analysis with DMAP1, Tip60, and MYC. Significantly, E1A 243R promotes association of MYC/MAX with the NuA4/Tip60 complex, implicating the importance of the MYC/NuA4 pathway in cellular transformation by both MYC and E1A.


Subject(s)
Adenovirus E1A Proteins/metabolism , Histone Acetyltransferases/metabolism , Multiprotein Complexes/metabolism , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-myc/metabolism , Adenovirus E1A Proteins/chemistry , Carrier Proteins/metabolism , Cell Line , Cell Transformation, Neoplastic/metabolism , Gene Expression , Genes, Reporter , Genetic Vectors/genetics , Humans , Lysine Acetyltransferase 5 , Models, Biological , Oncogene Proteins/metabolism , Protein Binding , Protein Interaction Mapping , Proteomics/methods , Proto-Oncogene Mas
16.
Genes Cancer ; 7(3-4): 98-109, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27382434

ABSTRACT

Human cancers frequently arise from increased expression of proto-oncogenes, such as MYC and HER2. Understanding the cellular pathways regulating the transcription and expression of proto-oncogenes is important for targeted therapies for cancer treatment. Adenoviral (Ad) E1A 243R (243 aa residues) is a viral oncoprotein that interacts with key regulators of gene transcription and cell proliferation. We have shown previously that the 80 amino acid N-terminal transcriptional repression domain of E1A 243R (E1A 1-80) can target the histone acetyltransferase (HAT) p300 and repress HER2 in the HER2-overexpressing human breast cancer cell line SKBR3. Expression of E1A 1-80 induces death of SKBR3 and other cancer cell lines. In this study, we performed total cell RNA sequence analysis and identified MYC as the regulatory gene for cellular proliferation most strongly repressed by E1A 1-80. By RT-quantitative PCR analysis we show that repression of MYC in SKBR3 cells occurs early after expression of E1A 1-80, suggesting that MYC may be an early responder of E1A 1-80-mediated transcriptional repression. Of interest, while E1A 1-80 repression of MYC occurs in all eight human cancer cell lines examined, repression of HER2 is cell-type dependent. We demonstrate by ChIP analysis that MYC transcriptional repression by E1A 1-80 is associated with inhibition of acetylation of H3K18 and H4K16 on the MYC promoter, as well as inhibition of RNA Pol II binding to the MYC promoter. Deletion mutant analysis of E1A 1-80 suggests that both p300/CBP and TRRAP are involved in E1A 1-80 repression of MYC transcription. Further, E1A 1-80 interaction with p300/CBP and TRRAP is correlated with inhibition of H3K18 and H4K16 acetylation on the MYC promoter, respectively. Our results indicate that E1A 1-80 may target two important pathways for histone modification to repress transcription in human cancer cells.

17.
Membranes (Basel) ; 5(4): 553-75, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26426064

ABSTRACT

Phosphatidylinositol 3-kinase-related kinases (PIKKs) play vital roles in the regulation of cell growth, proliferation, survival, and consequently metabolism, as well as in the cellular response to stresses such as ionizing radiation or redox changes. In humans six family members are known to date, namely mammalian/mechanistic target of rapamycin (mTOR), ataxia-telangiectasia mutated (ATM), ataxia- and Rad3-related (ATR), DNA-dependent protein kinase catalytic subunit (DNA-PKcs), suppressor of morphogenesis in genitalia-1 (SMG-1), and transformation/transcription domain-associated protein (TRRAP). All fulfill rather diverse functions and most of them have been detected in different cellular compartments including various cellular membranes. It has been suggested that the regulation of the localization of signaling proteins allows for generating a locally specific output. Moreover, spatial partitioning is expected to improve the reliability of biochemical signaling. Since these assumptions may also be true for the regulation of PIKK function, the current knowledge about the regulation of the localization of PIKKs at different cellular (membrane) compartments by a network of interactions is reviewed. Membrane targeting can involve direct lipid-/membrane interactions as well as interactions with membrane-anchored regulatory proteins, such as, for example, small GTPases, or a combination of both.

18.
Cell Cycle ; 13(10): 1583-9, 2014.
Article in English | MEDLINE | ID: mdl-24675885

ABSTRACT

The transformation/transcription domain-associated protein (TRRAP) is a common component of many histone acetyltransferase (HAT) complexes. Targeted-deletion of the Trrap gene led to early embryonic lethality and revealed a critical function of TRRAP in cell proliferation. Here, we investigate the function of TRRAP in murine B cells. To this end, we ablated Trrap gene in a B cell-restricted manner and studied its impact on B-cell development and proliferation, a pre-requisite for class switch recombination (CSR), the process that allows IgM-expressing B lymphocytes to switch to the expression of IgG, IgE, or IgA isotypes. We show that TRRAP deficiency impairs B-cell development but does not directly affect CSR. Instead, cells induced to proliferate undergo apoptosis. Our findings demonstrate a central and general role of TRRAP in cell proliferation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , B-Lymphocytes/metabolism , Histone Acetyltransferases/metabolism , Nuclear Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis , B-Lymphocytes/cytology , Cell Proliferation , Immunoglobulin Class Switching , Mice, Mutant Strains , Nuclear Proteins/metabolism , Organ Specificity
19.
Oncol Lett ; 6(2): 437-441, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24137342

ABSTRACT

Frequent somatic mutations in the GNA11, matrix metalloproteinase (MMP)27, FGD1, TRRAP and GRM3 genes have been reported in various types of human cancer, but whether these genes are mutated in thyroid cancer is not known. In the present study, a mutational analysis of these genes was performed in thyroid cancer cell lines and thyroid cancer samples. No GNA11 mutations were identified in the papillary thyroid cancer (PTC), follicular thyroid cancer (FTC) and anaplastic thyroid cancer (ATC) samples. Additionally, no mutations were identified in the MMP27 gene, although three synonymous [C351T (N117N), C1089T (S363S) and G1227A (G409G)] single nucleotide polymorphisms (SNPs) were observed infrequently in ATC. No mutations were detected in the FGD1 gene, but two infrequent synonymous [T2091C (T697T) and A2136G (P712P)] SNPs were observed in PTC. Furthermore, no mutations were identified in TRRAP and GRM3, although a frequent synonymous SNP [G1323A (T441T)] and infrequent non-synonymous SNP [G1424A (G475D)] of GRM3 were observed in PTC. No mutation of these genes was observed in 12 cell lines derived from various types of thyroid cancer. The present study reports for the first time the mutational status of the GNA11, MMP27, FGD1, TRRAP and GRM3 genes in thyroid cancer. No mutations were identified in these genes in the various types and cell lines of thyroid cancer. Therefore, unlike in other types of cancer, mutations in these genes are absent or uncommon in thyroid cancer.

20.
Gene ; 531(1): 112-6, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24001777

ABSTRACT

Ring finger protein 43 (RNF43) is an E3 ubiquitin-protein ligase that accepts ubiquitin from an E2 ubiquitin-conjugating enzyme and directly transfers the ubiquitin to targeted substrate proteins. Recently, large-scale sequencing efforts have identified prevalent RNF43 mutations in pancreatic and ovarian mucinous carcinomas. In the present study, we sequenced the entire coding sequences of RNF43 in 251 Chinese patients with distinct subtypes of ovarian cancers for the presence of RNF43 mutations. A total of 2 novel heterozygous nonsynonymous RNF43 mutations were identified in 2 out of 15 (13.3%) patients with mucinous ovarian carcinoma, these mutations were evolutionarily highly conserved; while no mutation was detected in other samples. In addition, none of the RNF43-mutated samples harbored DICER1 (dicer 1, ribonuclease type III), PPP2R1A (protein phosphatase 2, regulatory subunit A, alpha), TRRAP (transformation/transcription domain-associated protein) and DNMT3A (DNA (cytosine-5-)-methyltransferase 3 alpha) hot-spot mutations. Recurrent RNF43 mutations existed in mucinous ovarian carcinomas implicated that these mutations might play crucial roles in the tumorigenesis of these patients, while the absence of DICER1, PPP2R1A, TRRAP and DNMT3A hot-spot mutations suggested that these genetic alterations might not play synergistic roles with RNF43 mutations in these individuals. Additionally, the absence of RNF43 mutations in other subtypes of ovarian carcinoma implicated that RNF43 mutations might not be actively involved in the pathogenesis of these disorders.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , DNA-Binding Proteins/genetics , Mutation , Oncogene Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Adolescent , Adult , Aged , Amino Acid Sequence , Amino Acid Substitution , Asian People/genetics , Child , Child, Preschool , China , DNA Methyltransferase 3A , Female , Humans , Middle Aged , Molecular Sequence Data , Sequence Alignment , Ubiquitin-Protein Ligases , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL