ABSTRACT
The chemotherapeutic agent 5-fluorouracil (5-FU) remains the backbone of postoperative adjuvant treatment for gastric cancer. However, fewer than half of patients with gastric cancer benefit from 5-FU-based chemotherapies owing to chemoresistance and limited clinical biomarkers. Here, we identified the SNF2 protein Polo-like kinase 1-interacting checkpoint helicase (PICH) as a predictor of 5-FU chemosensitivity and characterized a transcriptional function of PICH distinct from its role in chromosome separation. PICH formed a transcriptional complex with RNA polymerase II (Pol II) and ATF4 at the CCNA1 promoter in an ATPase-dependent manner. Binding of the PICH complex promoted cyclin A1 transcription and accelerated S-phase progression. Overexpressed PICH impaired 5-FU chemosensitivity in human organoids and patient-derived xenografts. Furthermore, elevated PICH expression was negatively correlated with survival in postoperative patients receiving 5-FU chemotherapy. Together, these findings reveal an ATPase-dependent transcriptional function of PICH that promotes cyclin A1 transcription to drive 5-FU chemoresistance, providing a potential predictive biomarker of 5-FU chemosensitivity for postoperative patients with gastric cancer and prompting further investigation into the transcriptional activity of PICH. SIGNIFICANCE: PICH binds Pol II and ATF4 in an ATPase-dependent manner to form a transcriptional complex that promotes cyclin A1 expression, accelerates S-phase progression, and impairs 5-FU chemosensitivity in gastric cancer.
Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Cyclin A1 , DNA Helicases/metabolism , Fluorouracil/pharmacology , Adenosine Triphosphatases/therapeutic use , Polo-Like Kinase 1ABSTRACT
SMARCA4 (BRG1) and SMARCA2 (BRM) are the two paralogous ATPases of the SWI/SNF chromatin remodeling complexes frequently inactivated in cancers. Cells deficient in either ATPase have been shown to depend on the remaining counterpart for survival. Contrary to this paralog synthetic lethality, concomitant loss of SMARCA4/2 occurs in a subset of cancers associated with very poor outcomes. Here, we uncover that SMARCA4/2-loss represses expression of the glucose transporter GLUT1, causing reduced glucose uptake and glycolysis accompanied with increased dependency on oxidative phosphorylation (OXPHOS); adapting to this, these SMARCA4/2-deficient cells rely on elevated SLC38A2, an amino acid transporter, to increase glutamine import for fueling OXPHOS. Consequently, SMARCA4/2-deficient cells and tumors are highly sensitive to inhibitors targeting OXPHOS or glutamine metabolism. Furthermore, supplementation of alanine, also imported by SLC38A2, restricts glutamine uptake through competition and selectively induces death in SMARCA4/2-deficient cancer cells. At a clinically relevant dose, alanine supplementation synergizes with OXPHOS inhibition or conventional chemotherapy eliciting marked antitumor activity in patient-derived xenografts. Our findings reveal multiple druggable vulnerabilities of SMARCA4/2-loss exploiting a GLUT1/SLC38A2-mediated metabolic shift. Particularly, unlike dietary deprivation approaches, alanine supplementation can be readily applied to current regimens for better treatment of these aggressive cancers.
Subject(s)
Glutamine , Neoplasms , Humans , Glucose Transporter Type 1 , Adenosine Triphosphatases/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Dietary Supplements , DNA Helicases/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolismABSTRACT
Amyotrophic lateral sclerosis (ALS) is regarded as a fatal neurodegenerative disease that is featured by progressive damage of the upper and lower motor neurons. To date, over 45 genes have been found to be connected with ALS pathology. The aim of this work was to computationally identify unique sets of protein hydrolysate peptides that could serve as therapeutic agents against ALS. Computational methods which include target prediction, protein-protein interaction, and peptide-protein molecular docking were used. The results showed that the network of critical ALS-associated genes consists of ATG16L2, SCFD1, VAC15, VEGFA, KEAP1, KIF5A, FIG4, TUBA4A, SIGMAR1, SETX, ANXA11, HNRNPL, NEK1, C9orf72, VCP, RPSA, ATP5B, and SOD1 together with predicted kinases such as AKT1, CDK4, DNAPK, MAPK14, and ERK2 in addition to transcription factors such as MYC, RELA, ZMIZ1, EGR1, TRIM28, and FOXA2. The identified molecular targets of the peptides that support multi-metabolic components in ALS pathogenesis include cyclooxygenase-2, angiotensin I-converting enzyme, dipeptidyl peptidase IV, X-linked inhibitor of apoptosis protein 3, and endothelin receptor ET-A. Overall, the results showed that AGL, APL, AVK, IIW, PVI, and VAY peptides are promising candidates for further study. Future work would be needed to validate the therapeutic properties of these hydrolysate peptides by in vitro and in vivo approaches.
Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Humans , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Molecular Docking Simulation , NF-E2-Related Factor 2/metabolism , Peptides/pharmacology , Peptides/metabolism , Superoxide Dismutase-1/genetics , DNA Helicases/metabolism , RNA Helicases/metabolism , Multifunctional Enzymes/metabolism , Kinesins/metabolism , Flavoproteins/metabolismABSTRACT
The proper development of male and female gametophytes is critical for successful sexual reproduction and requires a carefully regulated series of events orchestrated by a suite of various proteins. RUVBL1 and RUVBL2, plant orthologues of human Pontin and Reptin, respectively, belong to the evolutionarily highly conserved AAA+ family linked to a wide range of cellular processes. Previously, we found that RUVBL1 and RUVBL2A mutations are homozygous lethal in Arabidopsis. Here, we report that RUVBL1 and RUVBL2A play roles in reproductive development. We show that mutant plants produce embryo sacs with an abnormal structure or with various numbers of nuclei. Although pollen grains of heterozygous mutant plants exhibit reduced viability and reduced pollen tube growth in vitro, some of the ruvbl pollen tubes are capable of targeting ovules in vivo. Similarly, some ruvbl ovules retain the ability to attract wild-type pollen tubes but fail to develop further. The activity of the RUVBL1 and RUVBL2A promoters was observed in the embryo sac, pollen grains, and tapetum cells and, for RUVBL2A, also in developing ovules. In summary, we show that the RUVBL proteins are essential for the proper development of both male and particularly female gametophytes in Arabidopsis.
Subject(s)
Arabidopsis Proteins , Arabidopsis , Humans , Germ Cells, Plant/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Pollen , Reproduction , Pollen Tube/genetics , Pollen Tube/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Helicases/genetics , DNA Helicases/metabolismABSTRACT
Stress granules (SGs) are cytoplasmic biomolecular condensates containing proteins and RNAs in response to stress. Ras-GTPase-activating protein binding protein 1 (G3BP1) is a core SG protein. Caprin-1 and ubiquitin specific peptidase 10 (USP10) interact with G3BP1, facilitating and suppressing SG formation, respectively. The crystal structures of the nuclear transport factor 2-like (NTF2L) domain of G3BP1 in complex with the G3BP1-interacting motif (GIM) of Caprin-1 and USP10 show that both GIMs bind to the same hydrophobic pocket of G3BP1. Moreover, both GIMs suppressed the liquid-liquid phase separation (LLPS) of G3BP1, suggesting that Caprin-1 likely facilitates SG formation via other mechanisms. Thus, we dissected various domains of Caprin-1 and investigated their role in LLPS in vitro and SG formation in cells. The C-terminal domain of Caprin-1 underwent spontaneous LLPS, whereas the N-terminal domain and GIM of Caprin-1 suppressed LLPS of G3BP1. The opposing effect of the N- and C-terminal domains of Caprin-1 on SG formation were demonstrated in cells with or without the endogenous Caprin-1. We propose that the N- and C-terminal domains of Caprin-1 regulate SG formation in a "yin and yang" fashion, mediating the dynamic and reversible assembly of SGs.
Subject(s)
DNA Helicases , RNA Helicases , RNA Recognition Motif Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , DNA Helicases/metabolism , Cytoplasmic Granules/metabolism , Stress Granules , GTPase-Activating Proteins/metabolism , Ubiquitin-Specific Proteases/metabolismABSTRACT
The prevalence of avian infectious bronchitis virus (IBV) is still one of causes inducing severe losses of production in the poultry industry worldwide. Vaccination does not completely prevent IBV infection and spread due to immune failure and viral mutations. ForsythiaeFructus and its compounds have been widely used in a lot of prescriptions of the traditional Chinese medicine for a long history, and it is well-known as safety and efficiency in heat-clearing and detoxifying. This study aims to investigate the anti-IBV activity and mechanism of phillygenin. The results showed that phillygenin inhibited IBV replication by disturbing multiple stages of the virus life cycle, including viral adsorption, invasion, internalization, and release in Vero cells. After being treated with 100, 125 and 150 µg/mL phillygenin, the expression of G3BP1 was significantly increased and the phosphorylation of PKR/eIF2α was activated, which increased stress granule, thereby triggering the antiviral response in Vero cells. The anti-virus activity of PHI was decreased when G3BP1 was interfered by si-RNA, and G3BP1 was down-regulated when PKR/eIF2α was interfered by si-RNA. In conclusion, our findings indicate that phillygenin activates PKR/eIF2α pathway and induces stress granule formation to exert anti-IBV, which holds promise to develop into a novel anti-IBV drug. Further study in vivo is needed to explore phillygenin as a potential and effective drug to prevent IB in poultry.
Subject(s)
Coronavirus Infections , Infectious bronchitis virus , Poultry Diseases , Animals , Chlorocebus aethiops , DNA Helicases/metabolism , DNA Helicases/pharmacology , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/pharmacology , Infectious bronchitis virus/physiology , Lignans , Poly-ADP-Ribose Binding Proteins , RNA , RNA Helicases/metabolism , RNA Helicases/pharmacology , RNA Recognition Motif Proteins , Stress Granules , Vero CellsABSTRACT
The bacterial single-stranded DNA-binding protein (SSB) uses an acidic C-terminal tail to interact with over a dozen proteins, acting as a genome maintenance hub. These SSB-protein interactions are essential, as mutations to the C-terminal tail that disrupt these interactions are lethal in Escherichia coli. While the roles of individual SSB-protein interactions have been dissected with mutational studies, small-molecule inhibitors of these interactions could serve as valuable research tools and have potential as novel antimicrobial agents. This chapter describes a high-throughput screening campaign used to identify inhibitors of SSB-protein interactions. A screen targeting the PriA-SSB interface from Klebsiella pneumoniae is presented as an example, but the methods may be adapted to target nearly any SSB interaction.
Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Klebsiella pneumoniae/metabolism , Small Molecule Libraries/pharmacology , Binding Sites , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Models, Molecular , Protein Binding/drug effects , Protein ConformationABSTRACT
INTRODUCTION: Dedifferentiated endometrial carcinoma is an uncommon highly aggressive uterine tumor. It comprises 2 components: a well-differentiated, low-grade epithelial carcinoma and an undifferentiated carcinoma. The undifferentiated carcinoma frequently exhibits rhabdoid cytologic features. Many of these tumors are characterized by an aberrant switch/sucrose non-fermenting (SWI/SNF) complex. They may also exhibit aberrant expression of mismatch repair (MMR) proteins. Together, these play an important role in the pathogenesis and aggressive nature of the tumor. MATERIAL AND METHODS: We present a case of dedifferentiated endometrial carcinoma in a 63-year-old female showing loss of expression of SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily A, member 4 (SMARCA4/BRG1), and aberrant expression of MMR proteins. We also review the literature starting from the earliest recognition of this entity and the various studies done to explain its molecular pathogenesis and prognostic importance. RESULTS AND CONCLUSIONS: Recognition of SWI/SNF complex-deficient dedifferentiated endometrial carcinoma is important as these tumors do not respond to platinum-based chemotherapy, and consideration of alternative therapies is often necessary. We also want to emphasize that though most of the studies have found MMR deficiency in the undifferentiated carcinoma component, it may be seen only in the low-grade, well-differentiated component, as observed in this case.
Subject(s)
Carcinoma/genetics , DNA Helicases/metabolism , Endometrial Neoplasms/genetics , Neoplasms, Complex and Mixed/genetics , Nuclear Proteins/metabolism , SMARCB1 Protein/metabolism , Transcription Factors/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/diagnosis , Carcinoma/drug therapy , Carcinoma/pathology , Cell Dedifferentiation/genetics , DNA Mismatch Repair , Drug Resistance, Neoplasm/genetics , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Endometrium/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Grading , Neoplasms, Complex and Mixed/diagnosis , Neoplasms, Complex and Mixed/drug therapy , Neoplasms, Complex and Mixed/pathologyABSTRACT
Paramutations represent directed and meiotically-heritable changes in gene regulation leading to apparent violations of Mendelian inheritance. Although the mechanism and evolutionary importance of paramutation behaviors remain largely unknown, genetic screens in maize (Zea mays) identify five components affecting 24 nucleotide RNA biogenesis as required to maintain repression of a paramutant purple plant1 (pl1) allele. Currently, the RNA polymerase IV largest subunit represents the only component also specifying proper development. Here we identify a chromodomain helicase DNA-binding 3 (CHD3) protein orthologous to Arabidopsis (Arabidopsis thaliana) PICKLE as another component maintaining both pl1 paramutation and normal somatic development but without affecting overall small RNA biogenesis. In addition, genetic tests show this protein contributes to proper male gametophyte function. The similar mutant phenotypes documented in Arabidopsis and maize implicate some evolutionarily-conserved gene regulation while developmental defects associated with the two paramutation mutants are largely distinct. Our results show that a CHD3 protein responsible for normal plant ontogeny and sperm transmission also helps maintain meiotically-heritable epigenetic regulatory variation for specific alleles. This finding implicates an intersection of RNA polymerase IV function and nucleosome positioning in the paramutation process.
Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Helicases/metabolism , Plant Proteins/metabolism , Pollen/metabolism , Zea mays/genetics , Alleles , Arabidopsis Proteins/genetics , DNA Helicases/genetics , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Plant , Genotype , Mutation , Phenotype , Phylogeny , Plant Proteins/genetics , Pollen/genetics , RNA, Plant/genetics , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Zea mays/growth & development , Zea mays/metabolismABSTRACT
Recent developments in chemotherapy focus on target-specific mechanisms, which occur only in cancer cells and minimize the effects on normal cells. DNA damage and repair pathways are a promising target in the treatment of cancer. In order to identify novel compounds targeting DNA repair pathways, two key proteins, 53BP1 and RAD54L, were tagged with fluorescent proteins as indicators for two major double strand break (DSB) repair pathways: non-homologous end-joining (NHEJ) and homologous recombination (HR). The engineered biosensor cells exhibited the same DNA repair properties as the wild type. The biosensor cells were further used to investigate the DNA repair activities of natural biological compounds. An extract from Phyllosticta sp., the endophyte isolated from the medicinal plant Garcinia cowa Roxb. ex Choisy, was tested. The results showed that the crude extract induced DSB, as demonstrated by the increase in the DNA DSB marker γH2AX. The damaged DNA appeared to be repaired through NHEJ, as the 53BP1 focus formation in the treated fraction was higher than in the control group. In conclusion, DNA repair-based biosensors are useful for the preliminary screening of crude extracts and biological compounds for the identification of potential targeted therapeutic drugs.
Subject(s)
Biosensing Techniques , DNA Damage , DNA Repair , Endophytes/chemistry , Garcinia/microbiology , Plant Extracts/pharmacology , Animals , Cell Line , Cell Survival , Chickens , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Fermentation , Fungi/metabolism , Garcinia/metabolism , Histones/metabolism , Homologous Recombination , Plants, Medicinal , Seeds/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolismABSTRACT
Astaxanthin (AST) is a product made from marine organisms that has been used as an anti-cancer supplement. It reduces pontin expression and induces apoptosis in SKBR3, a breast cancer cell line. Using Western blotting and qRT-PCR analyses, this study revealed that in the T47D and BT20 breast cancer cell lines, AST inhibits expression of pontin and mutp53, as well as the Oct4 and Nanog cancer stem cell (CSC) stemness genes. In addition, we explored the mechanism by which AST eradicates breast cancer cells using pontin siRNAs. Pontin knockdown by pontin siRNA reduced proliferation, Oct4 and Nanog expression, colony and spheroid formation, and migration and invasion abilities in breast cancer cells. In addition, reductions in Oct4, Nanog, and mutp53 expression following rottlerin treatment confirmed the role of pontin in these cells. Therefore, pontin may play a central role in the regulation of CSC properties and in cell proliferation following AST treatment. Taken together, these findings demonstrate that AST can repress CSC stemness genes in breast cancer cells, which implies that AST therapy could be used to improve the efficacy of other anti-cancer therapies against breast cancer cells.
Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Carrier Proteins/metabolism , DNA Helicases/metabolism , Neoplastic Stem Cells/drug effects , Tumor Suppressor Protein p53/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA Helicases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mutation , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Tumor Suppressor Protein p53/genetics , Xanthophylls/pharmacologyABSTRACT
The mechanism of unwinding catalyzed by the hepatitis C virus nonstructural protein 3 helicase (NS3h) has been a subject of considerable interest, with NS3h serving as a prototypical enzyme in the study of helicase function. Recent studies support an ATP-fueled, inchworm-like stepping of NS3h on the nucleic acid that would result in the displacement of the complementary strand of the duplex during unwinding. Here, we describe the screening of a site of incorporation of an unnatural amino acid in NS3h for fluorescent labeling of the enzyme to be used in single-molecule Förster resonance energy transfer (FRET) experiments. From the nine potential sites identified in NS3h for incorporation of the unnatural amino acid, only one allowed for expression and fluorescent labeling of the recombinant protein. Incorporation of the unnatural amino acid was confirmed via bulk assays to not interfere with unwinding activity of the helicase. Binding to four different dsDNA sequences bearing a ssDNA overhang segment of varying length (either minimal 6 or 7 base length overhang to ensure binding or a long 24 base overhang) and sequence was recorded with the new NS3h construct at the single-molecule level. Single-molecule fluorescence displayed time intervals with anticorrelated donor and acceptor emission fluctuations associated with protein binding to the substrates. An apparent FRET value was estimated from the binding events showing a single FRET value of â¼0.8 for the 6-7 base overhangs. A smaller mean value and a broad distribution was in turn recorded for the long ssDNA overhang, consistent with NS3h exploring a larger physical space while bound to the DNA construct. Notably, intervals where NS3h binding was recorded were exhibited at time periods where the acceptor dye reversibly bleached. Protein induced fluorescence intensity enhancement in the donor channel became apparent at these intervals. Overall, the site-specific fluorescent labeling of NS3h reported here provides a powerful tool for future studies to monitor the dynamics of enzyme translocation during unwinding by single-molecule FRET.
Subject(s)
Hepacivirus/enzymology , Single Molecule Imaging/methods , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Azides/chemistry , Binding Sites , DNA/metabolism , DNA Helicases/chemistry , DNA Helicases/metabolism , Escherichia coli/genetics , Fluorescence Resonance Energy Transfer , Genetic Code , Hepacivirus/genetics , Models, Molecular , Mutagenesis, Site-Directed , Phenylalanine/analogs & derivatives , Phenylalanine/chemistry , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Viral Nonstructural Proteins/chemistryABSTRACT
Galectin-3 and galectin-3 binding proteins (G3BP) are implicated as key players in metastasis. In the current study, we evaluated the effect of pectic polysaccharides on galectin-3 and G3BP mediated metastasis in vitro (cells) and in vivo (tissues). In vitro study (double immunostaining) confirms the presence of galectin-3 on the cell surface and G3BP in the interlinking region of the cells confirming the role of G3BP in bridging galectin-3 molecules. Dietary carrot (Daucus carota) pectic polysaccharide (CRPP) blocked the expression of galectin-3 and G3BP more effectively (80%), whereas the expressions were reduced to 60% upon treatment with swallow root (Decalepis hamiltonii) pectic polysaccharide (SRPP), ßcarotene and deferoxamine (antiproliferative drug). Ginger (Gingiber officinale) pectic polysaccharide (GRPP) showed only 20% reduction. CRPP reduced 80% of tumor incidence followed by cyclophosphamide - a chemotherapeutic drug (77%), SRPP (67%) and GRPP (45%). Further 3-5 folds reduction in galectin-3/G3BP expression followed by infiltration of macrophages into the deeper layer of the skin by CRPP and SRPP suggested the anticancer property via immunomodulation. Surface Plasmon Resonance (SPR) studies confirm galectin-3 and G3BP interaction, which are disrupted during the treatment with dietary pectic polysaccharides (DPP) (Supplementary Scheme-1). Overall data demonstrate the role of DPPs as potential anticancer alternatives.
Subject(s)
Apoptosis , DNA Helicases/metabolism , Dietary Carbohydrates/metabolism , Galectin 3/genetics , Galectin 3/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Polysaccharides/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blood Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Dietary Carbohydrates/pharmacology , Fluorescent Antibody Technique , Galectins , Gene Expression , Immunohistochemistry , Melanoma, Experimental , Mice , Polysaccharides/pharmacology , Skin Neoplasms/etiology , Skin Neoplasms/metabolismABSTRACT
OBJECTIVES: SMARCA4-deficient thoracic sarcoma(DTS) is a recently identified new entity of thoracic malignancies characterized by inactivation of SMARCA4. Patients with SMARCA4-DTS have a particulary aggresive clinical course and no effective treatments. However, the detailed clinical features of SMARCA4-DTS remain unclear. Here, we report the clinical courses and molecular profiles of two cases of SMARCA4-DTS. MATERIALS AND METHODS: We experienced strikingly similar two patients of SMARCA4-DTS. The clinicopathologic features were reviewed, and detailed immunohistochemical and comprehensive cancer panel analysis with next generation sequencing confirmed the diagnosis. RESULTS: Our cases had many clinical and radiological observations characteristic of SMARCA4-DTS in common. Immunohistochemical staing showed complete loss of SMARCA4 in tumor cells. Loss of function mutations were detected in SMARCA4. We found that severe SREs comprise a new significant clinical feature of SMARCA4-DTS. CONCLUSION: Integrated clinico-radiologic-pathologic-genetic diagnosis is essential for SMARCA4-DTS and physicians should pay attention to severe SREs during the clinical course of this disease.
Subject(s)
Bone Diseases/diagnosis , Bone and Bones/pathology , DNA Helicases/genetics , Lung/pathology , Mutation/genetics , Nuclear Proteins/genetics , Sarcoma/diagnosis , Thoracic Neoplasms/diagnosis , Transcription Factors/genetics , Biomarkers, Tumor/genetics , Bone Diseases/genetics , Bone Diseases/pathology , Bone and Bones/diagnostic imaging , DNA Helicases/metabolism , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Lung/diagnostic imaging , Male , Middle Aged , Nuclear Proteins/metabolism , Prognosis , Sarcoma/genetics , Sarcoma/pathology , Thoracic Neoplasms/genetics , Thoracic Neoplasms/pathology , Transcription Factors/metabolismABSTRACT
Osteosarcoma (OS) is an aggressive malignancy of bone affecting children, adolescents and young adults. Understanding vitamin D metabolism and vitamin D regulated genes in OS is an important aspect of vitamin D/cancer paradigm, and in evaluating vitamin D as adjuvant therapy for human OS. Vitamin D treatment of 143B OS cells induced significant and novel changes in the expression of genes that regulate: (a) inflammation and immunity; (b) formation of reactive oxygen species, metabolism of cyclic nucleotides, sterols, vitamins and mineral (calcium), quantity of gap junctions and skeletogenesis; (c) bone mineral density; and (d) cell viability of skeletal cells, aggregation of bone cancer cells and exocytosis of secretory vesicles. Ingenuity pathway analysis revealed significant reduction in Runx2 target genes such as fibroblast growth factor -1, -12 (FGF1 and FGF12), bone morphogenetic factor-1 (BMP1), SWI/SNF related, matrix associated actin dependent regulator of chromatin subfamily a, member 4 (SMARCA4), Matrix extracellular phosphoglycoprotein (MEPE), Integrin, ß4 (ITGBP4), Matrix Metalloproteinase -1, -28 (MMP1 and MMP28), and signal transducer and activator of transcription-4 (STAT4) in vitamin D treated 143B OS cells. These genes interact with the inflammation, oxidative stress and membrane vesicle biogenesis gene networks. Vitamin D not only inhibited the expression of Runx2 target genes MMP1, MMP28 and kallikrein related peptidase-7 (KLK7), but also migration and invasion of 143B OS cells. Vitamin D regulated Runx2 target genes or their products represent potential therapeutic targets and laboratory biomarkers for applications in translational oncology.
Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Gene Regulatory Networks , Osteosarcoma/metabolism , Oxidative Stress , Transport Vesicles/genetics , Vitamin D/pharmacology , Vitamins/pharmacology , Bone Morphogenetic Protein 1/genetics , Bone Morphogenetic Protein 1/metabolism , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Inflammation/genetics , Integrin beta4/genetics , Integrin beta4/metabolism , Kallikreins/genetics , Kallikreins/metabolism , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transport Vesicles/metabolismABSTRACT
Mutations in the Senataxin gene, SETX are known to cause the neurodegenerative disorders, ataxia with oculomotor apraxia type 2 (AOA2), and amyotrophic lateral sclerosis 4 (ALS4). However, the mechanism underlying disease pathogenesis is still unclear. The Senataxin N-terminal protein-interaction and C-terminal RNA/DNA helicase domains are conserved in the Saccharomyces cerevisiae homolog, Sen1p. Using genome-wide expression analysis, we first show alterations in key cellular pathways such as: redox, unfolded protein response, and TOR in the yeast sen1 ΔN mutant (N-terminal truncation). This mutant exhibited growth defects on nonfermentable carbon sources, was sensitive to oxidative stress, and showed severe loss of mitochondrial DNA. The growth defect could be partially rescued upon supplementation with reducing agents and antioxidants. Furthermore, the mutant showed higher levels of reactive oxygen species, lower UPR activity, and alterations in mitochondrial membrane potential, increase in vacuole acidity, free calcium ions in the cytosol, and resistance to rapamycin treatment. Notably, the sen1 ∆N mutant showed increased cell death and shortened chronological life span. Given the strong similarity of the yeast and human Sen1 proteins, our study thus provides a mechanism for the progressive neurological disorders associated with mutations in human senataxin.
Subject(s)
DNA Helicases/genetics , Mitochondria/genetics , Protein Serine-Threonine Kinases/genetics , RNA Helicases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Autophagy/genetics , Cardiolipins/biosynthesis , Cellular Senescence/genetics , DNA Helicases/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Fungal , Gene Regulatory Networks , Homeostasis/genetics , Humans , Immunoblotting , Membrane Potential, Mitochondrial/genetics , Microbial Viability/genetics , Microscopy, Fluorescence , Mitochondria/metabolism , Models, Genetic , Multifunctional Enzymes , Mutation , Oxidation-Reduction , Protein Serine-Threonine Kinases/metabolism , RNA Helicases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction/genetics , Unfolded Protein Response/geneticsABSTRACT
The BCCIP (BRCA2- and CDKN1A-interacting protein) is an important cofactor for BRCA2 in tumor suppression. Although the low expression of BCCIP is observed in multiple clinically diagnosed primary tumor tissues such as ovarian cancer, renal cell carcinoma and colorectal carcinoma, the mechanism of how BCCIP is regulated in cells is still unclear. The human INO80/YY1 chromatin remodeling complex composed of 15 subunits catalyzes ATP-dependent sliding of nucleosomes along DNA. Here, we first report that BCCIP is a novel target gene of the INO80/YY1 complex by presenting a series of experimental evidence. Gene expression studies combined with siRNA knockdown data locked candidate genes including BCCIP of the INO80/YY1 complex. Silencing or over-expressing the subunits of the INO80/YY1 complex regulates the expression level of BCCIP both in mRNA and proteins in cells. Also, the functions of INO80/YY1 complex in regulating the transactivation of BCCIP were confirmed by luciferase reporter assays. Chromatin immunoprecipitation (ChIP) experiments clarify the enrichment of INO80 and YY1 at +0.17 kb downstream of the BCCIP transcriptional start site. However, this enrichment is significantly inhibited by either knocking down INO80 or YY1, suggesting the existence of both INO80 and YY1 is required for recruiting the INO80/YY1 complex to BCCIP promoter region. Our findings strongly indicate that BCCIP is a potential target gene of the INO80/YY1 complex.
Subject(s)
Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly/physiology , DNA Helicases/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Transcription, Genetic/physiology , YY1 Transcription Factor/metabolism , ATPases Associated with Diverse Cellular Activities , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/genetics , DNA Helicases/genetics , DNA-Binding Proteins , HeLa Cells , Humans , Multiprotein Complexes/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic/physiology , YY1 Transcription Factor/geneticsABSTRACT
The presence of a highly conserved DNA binding domain in INO80 subfamily predicted that INO80 directly interacts with DNA and we demonstrated its DNA binding activity in vitro. Here we report the consensus motif recognized by the DBINO domain identified by SELEX method and demonstrate the specific interaction of INO80 with the consensus motif. We show that INO80 significantly down regulates the reporter gene expression through its binding motif, and the repression is dependent on the presence of INO80 but not YY1 in the cell. The interaction is lost if specific residues within the consensus motif are altered. We identify a large number of potential target sites of INO80 in the human genome through in silico analysis that can grouped into three classes; sites that contain the recognition sequence for INO80 and YY1, only YY1 and only INO80. We demonstrate the binding of INO80 to a representative set of sites in HEK cells and the correlated repressive histone modifications around the binding motif. In the light of the role of INO80 in homeotic gene regulation in Drosophila as an Enhancer of trithorax and polycomb protein (ETP) that can modify the effect of both repressive complexes like polycomb as well as the activating complex like trithorax, it remains to be seen if INO80 can act as a recruiter of chromatin modifying complexes.
Subject(s)
Chromatin/chemistry , DNA Helicases/metabolism , DNA/metabolism , Histones/metabolism , YY1 Transcription Factor/metabolism , ATPases Associated with Diverse Cellular Activities , Animals , Base Sequence , Binding Sites , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Chromatin/metabolism , Chromatin Assembly and Disassembly , Cloning, Molecular , DNA Helicases/antagonists & inhibitors , DNA Helicases/genetics , DNA-Binding Proteins , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HEK293 Cells , Histones/genetics , Humans , Protein Binding , Protein Interaction Domains and Motifs , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SELEX Aptamer Technique , Transcription Factors/genetics , Transcription Factors/metabolism , YY1 Transcription Factor/geneticsABSTRACT
Ying Yang 1 (YY1) is a ubiquitously expressed transcription factor shown to be essential for pro-B-cell development. However, the role of YY1 in other B-cell populations has never been investigated. Recent bioinformatics analysis data have implicated YY1 in the germinal center (GC) B-cell transcriptional program. In accord with this prediction, we demonstrated that deletion of YY1 by Cγ1-Cre completely prevented differentiation of GC B cells and plasma cells. To determine if YY1 was also required for the differentiation of other B-cell populations, we deleted YY1 with CD19-Cre and found that all peripheral B-cell subsets, including B1 B cells, require YY1 for their differentiation. Transitional 1 (T1) B cells were the most dependent upon YY1, being sensitive to even a half-dosage of YY1 and also to short-term YY1 deletion by tamoxifen-induced Cre. We show that YY1 exerts its effects, in part, by promoting B-cell survival and proliferation. ChIP-sequencing shows that YY1 predominantly binds to promoters, and pathway analysis of the genes that bind YY1 show enrichment in ribosomal functions, mitochondrial functions such as bioenergetics, and functions related to transcription such as mRNA splicing. By RNA-sequencing analysis of differentially expressed genes, we demonstrated that YY1 normally activates genes involved in mitochondrial bioenergetics, whereas it normally down-regulates genes involved in transcription, mRNA splicing, NF-κB signaling pathways, the AP-1 transcription factor network, chromatin remodeling, cytokine signaling pathways, cell adhesion, and cell proliferation. Our results show the crucial role that YY1 plays in regulating broad general processes throughout all stages of B-cell differentiation.
Subject(s)
B-Lymphocytes/physiology , Cell Differentiation , Gene Expression Regulation , Germinal Center/physiology , YY1 Transcription Factor/physiology , Animals , Cell Lineage , DNA Helicases/metabolism , Female , Germinal Center/cytology , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Mice, Inbred C57BLABSTRACT
The insulin-like growth factor-1 receptor (IGF-1R) is a receptor tyrosine kinase that has crucial roles in cell proliferation and protection from apoptosis. It is therefore not surprising that IGF-1R is often found overexpressed in many types of tumors. This has made IGF-1R a prominent target molecule for pharmacological companies to develop new anti-cancer agents. However, several clinical trials during the last 5 years using IGF-1R specific antibodies have shown disappointing results. We have previously shown that upon IGF-1 stimulation, the receptor becomes SUMOylated and translocates into the nucleus of cancer cells to act as a transcription co-factor. Soon after our original study, several others have reported nuclear IGF-1R (nIGF-1R) as well, and some of them have demonstrated a prognostic value of nIGF-1R expression in cancer. In the current study we demonstrate that nIGF-1R binds to and phosphorylates histone H3 at tyrosine 41 (H3Y41) in HeLa cells. Furthermore, our results suggest that phosphorylation of H3Y41 by nIGF-1R, stabilizes the binding of Brg1 chromatin remodeling protein to Histone H3. Our findings suggest that phosphorylated nIGF-1R, rather than total nIGF-1R, plays a superior role in these contexts. We identified SNAI2 oncogene as a target gene for nIGF-1R and its expression was decreased upon mutation of H3Y41 or by Brg1 knockdown. Furthermore, we demonstrate that both IGF-1R and Brg1 binds to the SNAI2 promoter. As SNAI2 protein is implicated in e.g. cancer invasion and metastasis, the nIGF-1R-mediated effects shown in this study may influence such important tumor phenotypic actions.