Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
1.
Genet Med ; 26(5): 101087, 2024 05.
Article in English | MEDLINE | ID: mdl-38288683

ABSTRACT

PURPOSE: Interneuronopathies are a group of neurodevelopmental disorders characterized by deficient migration and differentiation of gamma-aminobutyric acidergic interneurons resulting in a broad clinical spectrum, including autism spectrum disorders, early-onset epileptic encephalopathy, intellectual disability, and schizophrenic disorders. SP9 is a transcription factor belonging to the Krüppel-like factor and specificity protein family, the members of which harbor highly conserved DNA-binding domains. SP9 plays a central role in interneuron development and tangential migration, but it has not yet been implicated in a human neurodevelopmental disorder. METHODS: Cases with SP9 variants were collected through international data-sharing networks. To address the specific impact of SP9 variants, in silico and in vitro assays were carried out. RESULTS: De novo heterozygous variants in SP9 cause a novel form of interneuronopathy. SP9 missense variants affecting the glutamate 378 amino acid result in severe epileptic encephalopathy because of hypomorphic and neomorphic DNA-binding effects, whereas SP9 loss-of-function variants result in a milder phenotype with epilepsy, developmental delay, and autism spectrum disorder. CONCLUSION: De novo heterozygous SP9 variants are responsible for a neurodevelopmental disease. Interestingly, variants located in conserved DNA-binding domains of KLF/SP family transcription factors may lead to neomorphic DNA-binding functions resulting in a combination of loss- and gain-of-function effects.


Subject(s)
Autism Spectrum Disorder , Epilepsy , Intellectual Disability , Interneurons , Sp Transcription Factors , Transcription Factors , Adolescent , Child , Child, Preschool , Female , Humans , Male , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Epilepsy/genetics , Epilepsy/pathology , Heterozygote , Intellectual Disability/genetics , Intellectual Disability/pathology , Interneurons/metabolism , Interneurons/pathology , Mutation, Missense/genetics , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Phenotype , Transcription Factors/genetics , Transcription Factors/metabolism , Sp Transcription Factors/genetics
2.
Int J Mol Sci ; 24(6)2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36982239

ABSTRACT

The specificity protein (Sp) transcription factors (TFs) Sp1, Sp2, Sp3 and Sp4 exhibit structural and functional similarities in cancer cells and extensive studies of Sp1 show that it is a negative prognostic factor for patients with multiple tumor types. In this review, the role of Sp1, Sp3 and Sp4 in the development of cancer and their regulation of pro-oncogenic factors and pathways is reviewed. In addition, interactions with non-coding RNAs and the development of agents that target Sp transcription factors are also discussed. Studies on normal cell transformation into cancer cell lines show that this transformation process is accompanied by increased levels of Sp1 in most cell models, and in the transformation of muscle cells into rhabdomyosarcoma, both Sp1 and Sp3, but not Sp4, are increased. The pro-oncogenic functions of Sp1, Sp3 and Sp4 in cancer cell lines were studied in knockdown studies where silencing of each individual Sp TF decreased cancer growth, invasion and induced apoptosis. Silencing of an individual Sp TF was not compensated for by the other two and it was concluded that Sp1, Sp3 and Sp4 are examples of non-oncogene addicted genes. This conclusion was strengthened by the results of Sp TF interactions with non-coding microRNAs and long non-coding RNAs where Sp1 contributed to pro-oncogenic functions of Sp/non-coding RNAs. There are now many examples of anticancer agents and pharmaceuticals that induce downregulation/degradation of Sp1, Sp3 and Sp4, yet clinical applications of drugs specifically targeting Sp TFs are not being used. The application of agents targeting Sp TFs in combination therapies should be considered for their potential to enhance treatment efficacy and decrease toxic side effects.


Subject(s)
Antineoplastic Agents , MicroRNAs , Rhabdomyosarcoma , Humans , Sp Transcription Factors/metabolism , Antineoplastic Agents/pharmacology , MicroRNAs/genetics , Rhabdomyosarcoma/genetics , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic
3.
Int J Mol Sci ; 23(17)2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36077352

ABSTRACT

Tumor development and progression depend on reprogramming of signaling pathways that regulate cell metabolism. Alterations to various metabolic pathways such as glycolysis, oxidative phosphorylation, lipid metabolism, and hexosamine biosynthesis pathway are crucial to sustain increased redox, bioenergetic, and biosynthesis demands of a tumor cell. Transcription factors (oncogenes and tumor suppressors) play crucial roles in modulating these alterations, and their functions are tethered to major metabolic pathways under homeostatic conditions and disease initiation and advancement. Specificity proteins (SPs) and Krüppel-like factors (KLFs) are closely related transcription factors characterized by three highly conserved zinc fingers domains that interact with DNA. Studies have demonstrated that SP and KLF transcription factors are expressed in various tissues and regulate diverse processes such as proliferation, differentiation, apoptosis, inflammation, and tumorigenesis. This review highlights the role of SP and KLF transcription factors in the metabolism of various cancers and their impact on tumorigenesis. A better understanding of the role and underlying mechanisms governing the metabolic changes during tumorigenesis could provide new therapeutic opportunities for cancer treatment.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Neoplasms/metabolism , Sp Transcription Factors/metabolism , Cell Transformation, Neoplastic , Humans , Zinc Fingers
4.
World J Gastroenterol ; 27(38): 6387-6398, 2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34720529

ABSTRACT

Specificity protein (Sp) transcription factors (TFs) Sp1, Sp3 and Sp4, and the orphan nuclear receptor 4A1 (NR4A1) are highly expressed in pancreatic tumors and Sp1 is a negative prognostic factor for pancreatic cancer patient survival. Results of knockdown and overexpression of Sp1, Sp3 and Sp4 in pancreatic and other cancer lines show that these TFs are individually pro-oncogenic factors and loss of one Sp TF is not compensated by other members. NR4A1 is also a pro-oncogenic factor and both NR4A1 and Sp TFs exhibit similar functions in pancreatic cancer cells and regulate cell growth, survival, migration and invasion. There is also evidence that Sp TFs and NR4A1 regulate some of the same genes including survivin, epidermal growth factor receptor, PAX3-FOXO1, α5- and α6-integrins, ß1-, ß3- and ß4-integrins; this is due to NR4A1 acting as a cofactor and mediating NR4A1/Sp1/4-regulated gene expression through GC-rich gene promoter sites. Several studies show that drugs targeting Sp downregulation or NR4A1 antagonists are highly effective inhibitors of Sp/NR4A1-regulated pathways and genes in pancreatic and other cancer cells, and the triterpenoid celastrol is a novel dual-acting agent that targets both Sp TFs and NR4A1.


Subject(s)
Pancreatic Neoplasms , Transcription Factors , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Oncogenes , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Sp Transcription Factors/genetics , Sp Transcription Factors/metabolism , Transcription Factors/genetics
5.
Invest Ophthalmol Vis Sci ; 61(8): 41, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32721022

ABSTRACT

Purpose: ZEB1 is induced during endothelial-mesenchymal transition (EnMT) in the cornea. Induction of SP1 and SP3 by ZEB1 along with identification of putative SP1 and SP3 binding sites in promoters of EnMT-associated gene lead us to investigate their roles in retrocorneal membrane formation in the corneal endothelium. Methods: Expressions of SP1, SP3, and EnMT associated genes were analyzed by immunoblotting and semiquantitative reverse transcription polymerase chain reaction. Accell SMARTpool siRNAs targeting ZEB1, SP1, and SP3 were used for gene knockdown. SP1 and SP3 binding to promoters of EnMT associated genes was investigated by chromatin immunoprecipitation assay. Corneal endothelium in mice was surgically injured in vivo under direct visualization. Results: Transient Fibroblast Growth Factor 2 stimulation increased the expression of both SP1 and SP3 in the human corneal endothelium ex vivo. ZEB1 siRNA knockdown inhibited FGF2-induced SP1 mRNA and protein but not the expression of SP3. FGF2-induced expression of EnMT-related genes, such as fibronectin, vimentin, and type I collagen, was reduced by both SP1 and SP3 siRNA knockdown, with inhibition of SP1 having a greater inhibitory effect than SP3. Additionally, although SP1 and SP3 proteins were found to bind together, SP1 and SP3 could bind to the same promoter binding sites of EnMT-related genes in the absence of the other. Moreover, siRNA knockdown of Zeb1 inhibited injury-dependent RCM formation in mouse corneal endothelium in vivo. Conclusions: Zeb1, through SP1 and SP3, plays a central role in mesenchymal transition induced fibrosis in the corneal endothelium and suggests that Zeb1 could be targeted to inhibit anterior segment fibrosis.


Subject(s)
Endothelium, Corneal , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Cells, Cultured , Endothelium, Corneal/metabolism , Endothelium, Corneal/pathology , Epithelial-Mesenchymal Transition/physiology , Fibrosis/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Mice , RNA, Small Interfering , Sp Transcription Factors/genetics , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/genetics , Zinc Fingers
6.
Dev Genes Evol ; 230(3): 239-245, 2020 05.
Article in English | MEDLINE | ID: mdl-32430690

ABSTRACT

The Sp-family genes encode important transcription factors in animal development. Here we investigate the embryonic expression patterns of the complete set of Sp-genes in the velvet worm Euperipatoides kanangrensis (Onychophora), with a special focus on the Sp6-9 ortholog. In arthropods, Sp6-9, the ortholog of the Drosophila melanogaster D-Sp1 gene plays a conserved role in appendage development. Our data show that the expression of Sp6-9 during the development of the velvet worm is conserved, suggesting that the key function of the Sp6-9 gene dates back to at least the last common ancestor of arthropods and onychophorans and thus likely the last common ancestor of Panarthropoda.


Subject(s)
Body Patterning/genetics , Invertebrates/embryology , Invertebrates/genetics , Sp Transcription Factors/genetics , Sp Transcription Factors/metabolism , Amino Acid Sequence , Animals , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Phylogeny , Zinc Fingers
7.
Int J Biol Sci ; 16(9): 1536-1550, 2020.
Article in English | MEDLINE | ID: mdl-32226300

ABSTRACT

Non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) wild-type is intrinsic resistance to EGFR-tyrosine kinase inhibitors (TKIs). In this study, we assessed whether the combination of bisdemethoxycurcumin (BDMC) and icotinib could surmount primary EGFR-TKI resistance in NSCLC cells and investigated its molecular mechanism. Results showed that the combination of BDMC and icotinib produced potently synergistic growth inhibitory effect on primary EGFR-TKI-resistant NSCLC cell lines H460 (EGFR wild-type and K-ras mutation) and H1781 (EGFR wild-type and Her2 mutation). Compared with BDMC or icotinib alone, the two drug combination induced more significant apoptosis and autophagy via suppressing EGFR activity and interaction of Sp1 and HDCA1/HDCA2, which was accompanied by accumulation of reactive oxygen species (ROS), induction of DNA damage, and inhibition of cell migration and invasion. ROS inhibitor (NAC) and autophagy inhibitors (CQ or 3-MA) partially reversed BDMC plus icotinib-induced growth inhibitory effect on the NSCLC cells. Meanwhile, co-treatment with NAC attenuated the two drug combination-induced autophagy, apoptosis, DNA damage and decrease of cell migration and invasion ability. Also, 3-MA or CQ can abate the combination treatment-induced apoptosis and DNA damage, suggesting that there is crosstalk between different signaling pathways in the effect produced by the combination treatment. Our data indicate that BMDC has the potential to improve the treatment of primary EGFR-TKI resistant NISCLC that cannot be controlled with single-target agent, such as icotinib.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Crown Ethers/therapeutic use , Diarylheptanoids/therapeutic use , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , DNA Damage , Drug Resistance, Neoplasm/drug effects , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Reactive Oxygen Species/metabolism , Sp Transcription Factors/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/antagonists & inhibitors
8.
Crit Rev Oncog ; 25(4): 355-363, 2020.
Article in English | MEDLINE | ID: mdl-33639062

ABSTRACT

Specificity protein (Sp) transcription factors regulate the expression of genes associated with several cellular processes and play a critical role in early development. Typically, Sp protein expression decreases with age in healthy adults. Research has shown that Sp proteins can impact the development and transformation of cancer cells and other oncogenic processes, including survival, proliferation, spread, and metastasis. Among the Sp proteins, Sp1, Sp3, and Sp4 have been the main targets of study as they are shown to be highly expressed in cancer cells compared to healthy cells. Increased levels of Sp1 are correlated with poor prognosis in some malignancies, including gastrointestinal cancers. In this review, we discuss the role of Sp transcription factors and examine their activities as pro-oncogenic factors in esophageal cancer (EC). Other aspects presented in this review are potential therapeutic options for EC that target Sp1. We summarize the published information on preclinical results using mithramycin and tolfenamic acid.


Subject(s)
Esophageal Neoplasms , Sp Transcription Factors , Carcinogenesis , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/genetics , Esophageal Neoplasms/therapy , Humans , Plicamycin/pharmacology , Prognosis , Risk Factors , Sp Transcription Factors/genetics , ortho-Aminobenzoates/pharmacology
9.
Cells ; 8(3)2019 03 07.
Article in English | MEDLINE | ID: mdl-30866413

ABSTRACT

Chicken ovalbumin upstream promoter-transcription factor I (COUP-TFI) is an orphan receptor and member of the nuclear receptor superfamily. Among a series of methylene substituted diindolylmethanes (C-DIMs) containing substituted phenyl and heteroaromatic groups, we identified 1,1-bis(3'-indolyl)-1-(4-pyridyl)-methane (DIM-C-Pyr-4) as an activator of COUP-TFI. Structure activity studies with structurally diverse heteroaromatic C-DIMs showed that the pyridyl substituted compound was active and the 4-pyridyl substituent was more potent than the 2- or 3-pyridyl analogs in transactivation assays in breast cancer cells. The DIM-C-Pyr-4 activated chimeric GAL4-COUP-TFI constructs containing full length, C- or N-terminal deletions, and transactivation was inhibited by phosphatidylinositol-3-kinase and protein kinase A inhibitors. However, DIM-C-Pyr-4 also induced transactivation and interactions of COUP-TFI and steroid receptor coactivators-1 and -2 in mammalian two-hybrid assays, and ligand-induced interactions of the C-terminal region of COUP-TFI were not affected by kinase inhibitors. We also showed that DIM-C-Pyr-4 activated COUP-TFI-dependent early growth response 1 (Egr-1) expression and this response primarily involved COUP-TFI interactions with Sp3 and to a lesser extent Sp1 bound to the proximal region of the Egr-1 promoter. Modeling studies showed interactions of DIM-C-Pyr-4 within the ligand binding domain of COUP-TFI. This report is the first to identify a COUP-TFI agonist and demonstrate activation of COUP-TFI-dependent Egr-1 expression.


Subject(s)
COUP Transcription Factor I/metabolism , Indoles/pharmacology , Animals , COUP Transcription Factor I/chemistry , Cell Line, Tumor , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Humans , Ligands , Mice , Models, Molecular , Nuclear Receptor Co-Repressor 2/metabolism , Sp Transcription Factors/metabolism
10.
Mol Cancer Res ; 17(3): 794-805, 2019 03.
Article in English | MEDLINE | ID: mdl-30610105

ABSTRACT

Methyl 2-trifluoromethyl-3,11-dioxo-18ß-olean-1,12-dien-3-oate (CF3DODA-Me) is derived synthetically from glycyrrhetinic acid, a major component of licorice, and this compound induced reactive oxygen species (ROS) in RD and Rh30 rhabdomyosarcoma (RMS) cells. CF3DODA-Me also inhibited growth and invasion and induced apoptosis in RMS cells, and these responses were attenuated after cotreatment with the antioxidant glutathione, demonstrating the effective anticancer activity of ROS in RMS. CF3DODA-Me also downregulated expression of specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4 and prooncogenic Sp-regulated genes including PAX3-FOXO1 (in Rh30 cells). The mechanism of CF3DODA-Me-induced Sp-downregulation involved ROS-dependent repression of c-Myc and cMyc-regulated miR-27a and miR-17/20a, and this resulted in induction of the miRNA-regulated Sp repressors ZBTB4, ZBTB10, and ZBTB34. The cell and tumor growth effects of CF3DODA-Me further emphasize the sensitivity of RMS cells to ROS inducers and their potential clinical applications for treating this deadly disease. IMPLICATIONS: CF3DODA-Me and HDAC inhibitors that induce ROS-dependent Sp downregulation could be developed for clinical applications in treating rhabdomyosarcoma.


Subject(s)
Reactive Oxygen Species/metabolism , Rhabdomyosarcoma/drug therapy , Sp Transcription Factors/genetics , Triterpenes/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Rhabdomyosarcoma/pathology , Transfection , Triterpenes/pharmacology
11.
Forensic Sci Int Genet ; 38: 1-8, 2019 01.
Article in English | MEDLINE | ID: mdl-30300865

ABSTRACT

Many studies have reported age-associated DNA methylation changes and age-predictive models in various tissues and body fluids. Although age-associated DNA methylation changes can be tissue-specific, a multi-tissue age predictor that is applicable to various tissues and body fluids with considerable prediction accuracy might be valuable. In this study, DNA methylation at 5 CpG sites from the ELOVL2, FHL2, KLF14, C1orf132/MIR29B2C, and TRIM59 genes were investigated in 448 samples from blood, saliva, and buccal swabs. A multiplex methylation SNaPshot assay was developed to measure DNA methylation simultaneously at the 5 CpG sites. Among the 5 CpG sites, 3 CpG sites in the ELOVL2, KLF14 and TRIM59 genes demonstrated strong correlation between DNA methylation and age in all 3 sample types. Age prediction models built separately for each sample type using the DNA methylation values at the 5 CpG sites showed high prediction accuracy with a Mean Absolute Deviation from the chronological age (MAD) of 3.478 years in blood, 3.552 years in saliva and 4.293 years in buccal swab samples. A tissue-combined model constructed with 300 training samples including 100 samples from each blood, saliva and buccal swab samples demonstrated a very strong correlation between predicted and chronological ages (r = 0.937) and a high prediction accuracy with a MAD of 3.844 years in the 148 independent test set samples of 50 blood, 50 saliva and 48 buccal swab samples. Although more validation might be needed, the tissue-combined model's prediction accuracies in each sample type were very much similar to those obtained from each tissue-specific model. The multiplex methylation SNaPshot assay and the age prediction models in our study would be useful in forensic analysis, which frequently involves DNA from blood, saliva, and buccal swab samples.


Subject(s)
Aging/genetics , Blood Chemical Analysis , DNA Methylation , Mouth Mucosa/chemistry , Saliva/chemistry , Acetyltransferases/genetics , Adolescent , Adult , Aged , CpG Islands/genetics , Fatty Acid Elongases , Forensic Genetics , Genetic Markers , Genotyping Techniques/instrumentation , Humans , Intracellular Signaling Peptides and Proteins , Kruppel-Like Transcription Factors , LIM-Homeodomain Proteins/genetics , Membrane Proteins/genetics , Metalloproteins/genetics , Middle Aged , Muscle Proteins/genetics , Sequence Analysis, DNA , Sp Transcription Factors/genetics , Transcription Factors/genetics , Tripartite Motif Proteins , Young Adult
12.
Atherosclerosis ; 278: 39-48, 2018 11.
Article in English | MEDLINE | ID: mdl-30248551

ABSTRACT

BACKGROUND AND AIMS: Human genetic studies indicated that variations near the transcription factor Krüppel-like factor 14 (KLF14) gene locus are highly associated with coronary artery disease. Activation of endothelial cells (ECs) by pro-inflammatory molecules and pathways is a primary step in atherosclerosis development. We aimed to investigate the effects and mechanism of KLF14 on inflammatory responses in ECs. METHODS: Adenovirus-mediated overexpression of human KLF14 and EC specific Klf14 knockout mice were applied to study the role of KLF14 in EC inflammation. Intravital microscopy was used to examine leukocyte-endothelial cell interactions in vivo. RESULTS: The expression of Klf14 was markedly decreased in mouse aortic ECs in both acute and chronic inflammatory conditions. Overexpression of KLF14 inhibited inflammatory activation of human ECs stimulated by interleukin 1ß and tumor necrosis factor α. Primary pulmonary ECs from Klf14 knockout mice showed increased expression of adhesion molecules under IL-1ß stimuli. Mechanistically, KLF14 inhibited NF-κB signaling pathway by transcriptionally suppressing the expression of p65, resulting in significantly decreased leukocyte adhesion to activated ECs. Using intravital microscopy, an increased leukocyte-endothelial cell interaction was observed in endothelial specific Klf14 knockout mice compared to wild type control mice. Additionally, perhexiline, a KLF14 activator, induces KLF14 expression in ECs and reduced leukocyte-endothelial cell interactions in vitro and in vivo. CONCLUSIONS: The data revealed that KLF14 inhibited the inflammatory response in ECs and the protective effects were mediated by transcriptional inhibition of NF-κB signaling pathway. Endothelial KLF14 could be a potential therapeutic target for cardiovascular diseases.


Subject(s)
Coronary Artery Disease/metabolism , Endothelium, Vascular/pathology , Inflammation/pathology , Sp Transcription Factors/metabolism , Transcription Factor RelA/metabolism , Animals , Down-Regulation , Human Umbilical Vein Endothelial Cells , Humans , Kruppel-Like Transcription Factors/metabolism , Leukocytes/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
13.
J Cell Physiol ; 234(1): 572-580, 2018 01.
Article in English | MEDLINE | ID: mdl-30216442

ABSTRACT

Long non-coding RNAs (lncRNAs) are a class of regulatory noncoding RNAs. Emerging evidence highlights the critical roles of lncRNAs in the progression of hepatocellular carcinoma (HCC). Although many lncRNAs have been identified in the development of HCC, the association between DiGeorge syndrome critical region gene 5 (DGCR5) and HCC remains unclear. In the current study, we focused on the biological role of DGCR5 in HCC. We observed that DGCR5 was decreased in HCC cells, including SMCC7721, Hep3B, HepG2, MHCC-97L, MHCC-97H, and SNU449 hepatocellular carcinoma cells, compared with the normal human liver cell line THLE-3 normal human liver cells. In addition, DGCR5 overexpression could repress HCC cell growth, migration, and invasion considerably. Increasing studies have indicated the interactions between lncRNAs and microRNAs. MicroRNAs are endogenous small noncoding RNAs and they can play important roles in tumorigenesis. MicroRNA 346 (miR-346) has been demonstrated in various human cancer types, including HCC. MiR-346 was found to be increased in HCC cells and DGCR5 can act as a sponge of miR-346 to modulate the progression of HCC. The binding correlation between DGCR5 and miR-346 was validated in our research. Subsequently, Krüppel-like factor 14 (KLF14) was predicted as a downstream target of miR-346 and miR-346 can induce the development of HCC by inhibiting KLF14. Finally, we proved that DGCR5 can rescue the inhibited levels of KLF14 repressed by miR-346 mimics in MHCC-97H and Hep3B cells. Taken together, it was indicated in our study that DGCR5 can restrain the progression of HCC through sponging miR-346 and modulating KLF14 in vitro.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Sp Transcription Factors/genetics , Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Hep G2 Cells , Humans , Kruppel-Like Transcription Factors , Liver Neoplasms/pathology
14.
Biochem Biophys Res Commun ; 503(3): 1848-1853, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30078677

ABSTRACT

Long noncoding RNAs (lncRNAs) represent a novel type of noncoding RNAs of over 200 nucleotides, characterized by no or limited protein-coding potential. Although the function of lncRNAs attracts increasing attention recently, the relationship between lncRNA and colorectal cancer (CRC) remains further investigation. In our study, we found that lncRNA HAND2-AS1 was markedly downregulated in CRC tissues. And its expression level was negatively correlated with metastasis and advanced stage in CRC patients. Furthermore, we showed that HAND2-AS1 low expression predicted poor prognosis. Functionally, we found that overexpression of HAND2-AS1 obviously attenuated the proliferation and invasion of CRC cells. Ectopic expression of HAND2-AS1 also inhibited tumor propagation in vivo. In mechanism, HAND2-AS1 served as a sponge of miR-1275 which targeted KLF14. Through facilitating KLF14 expression, HAND2-AS1 suppressed CRC progression. In conclusion, our study demonstrated that HAND2-AS1 exerts a suppressive role in CRC by sponging miR-1275 and modulating KLF14 expression.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Sp Transcription Factors/biosynthesis , Animals , Cell Proliferation , Cells, Cultured , Colorectal Neoplasms/metabolism , Disease Progression , Humans , Kruppel-Like Transcription Factors , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , RNA, Long Noncoding/genetics , Sp Transcription Factors/genetics , Sp Transcription Factors/metabolism , Up-Regulation
15.
Mol Pharmacol ; 94(4): 1187-1196, 2018 10.
Article in English | MEDLINE | ID: mdl-30115673

ABSTRACT

Bortezomib alone and in combination with other anticancer agents are extensively used for chemotherapeutic treatment of multiple myeloma (MM) patients and are being developed for treating other cancers. Bortezomib acts through multiple pathways, and in this study with ANBL-6 and RPMI 8226 MM cells we show that bortezomib inhibited growth and induced apoptosis and that this was accompanied by downregulation of specificity protein (Sp) 1, Sp3, and Sp4 transcription factors that are overexpressed in these cells. Similar results were observed in pancreatic and colon cancer cells. The functional importance of this pathway was confirmed by showing that individual knockdown of Sp1, Sp3, and Sp4 in MM cells inhibited cell growth and induced apoptosis, and that this correlates with the results of previous studies in pancreatic, colon, and other cancer cell lines. The mechanism of bortezomib-mediated downregulation of Sp transcription factors in MM was due to the induction of caspase-8 and upstream factors, including Fas-associated death domain. These results demonstrate that an important underlying mechanism of action of bortezomib was due to the activation of caspase-8-dependent downregulation of Sp1, Sp3, Sp4, and pro-oncogenic Sp-regulated genes.


Subject(s)
Bortezomib/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Sp Transcription Factors/metabolism , Apoptosis/drug effects , Caspase 8/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Fas-Associated Death Domain Protein/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism
16.
Oxid Med Cell Longev ; 2018: 6918797, 2018.
Article in English | MEDLINE | ID: mdl-29849909

ABSTRACT

Epigenetic mechanisms play an important role in the development and progression of various neurodegenerative diseases. Abnormal methylation of numerous genes responsible for regulation of transcription, DNA replication, and apoptosis has been linked to Alzheimer's disease (AD) pathology. We have recently performed whole transcriptome profiling of familial early-onset Alzheimer's disease (fEOAD) patient-derived fibroblasts. On this basis, we demonstrated a strong dysregulation of cell cycle checkpoints and DNA damage response (DDR) in both fibroblasts and reprogrammed neurons. Here, we show that the aging-correlated hypermethylation of KLF14 and TRIM59 genes associates with abnormalities in DNA repair and cell cycle control in fEOAD. Based on the resulting transcriptome networks, we found that the hypermethylation of KLF14 might be associated with epigenetic regulation of the chromatin organization and mRNA processing followed by hypermethylation of TRIM59 likely associated with the G2/M cell cycle phase and p53 role in DNA repair with BRCA1 protein as the key player. We propose that the hypermethylation of KLF14 could constitute a superior epigenetic mechanism for TRIM59 hypermethylation. The methylation status of both genes affects genome stability and might contribute to proapoptotic signaling in AD. Since this study combines data obtained from various tissues from AD patients, it reinforces the view that the genetic methylation status in the blood may be a valuable predictor of molecular processes occurring in affected tissues. Further research is necessary to define a detailed role of TRIM59 and KLF4 in neurodegeneration of neurons.


Subject(s)
Alzheimer Disease/pathology , DNA Methylation , Membrane Proteins/metabolism , Metalloproteins/metabolism , Signal Transduction , Sp Transcription Factors/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Apoptosis , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Cycle Checkpoints , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , DNA Repair , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Regulatory Networks , Humans , Intracellular Signaling Peptides and Proteins , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Male , Membrane Proteins/genetics , Metalloproteins/genetics , Middle Aged , Sp Transcription Factors/genetics , Tripartite Motif Proteins , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Genome Biol ; 19(1): 74, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29880058

ABSTRACT

We discuss a recent study that has identified and validated the link between a type-2 diabetes (T2D) association and human adipose biology by means of KLF14 gene expression. In addition to being maternally imprinted, the contributed risk at this locus is greater in female carriers.


Subject(s)
Adiposity/genetics , Genetic Loci/genetics , Genetic Predisposition to Disease/genetics , Animals , Diabetes Mellitus, Type 2/genetics , Gene Expression , Genome-Wide Association Study/methods , Genomics/methods , Humans , Sp Transcription Factors/genetics
18.
Sci Rep ; 8(1): 6374, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29686323

ABSTRACT

Curly coat represents an extraordinary type of coat in horses, particularly seen in American Bashkir Curly Horses and Missouri Foxtrotters. In some horses with curly coat, a hypotrichosis of variable extent was observed, making the phenotype appear more complex. In our study, we aimed at investigating the genetic background of curly coat with and without hypotrichosis using high density bead chip genotype and next generation sequencing data. Genome-wide association analysis detected significant signals (p = 1.412 × 10-05-1.102 × 10-08) on horse chromosome 11 at 22-35 Mb. In this significantly associated region, six missense variants were filtered out from whole-genome sequencing data of three curly coated horses of which two variants within KRT25 and SP6 could explain all hair phenotypes. Horses heterozygous or homozygous only for KRT25 variant showed curly coat and hypotrichosis, whereas horses with SP6 variant only, exhibited curly coat without hypotrichosis. Horses with mutant alleles in both variants developed curly hair and hypotrichosis. Thus, mutant KRT25 allele is masking SP6 allele effect, indicative for epistasis of KRT25 variant over SP6 variant. In summary, genetic variants in two different genes, KRT25 and SP6, are responsible for curly hair. All horses with KRT25 variant are additionally hypotrichotic due to the KRT25 epistatic effect on SP6.


Subject(s)
Animal Fur/chemistry , Epistasis, Genetic , Horses/genetics , Hypotrichosis/genetics , Keratins, Hair-Specific/genetics , Polymorphism, Single Nucleotide , Sp Transcription Factors/genetics , Animals , Chromosomes, Mammalian , Genome-Wide Association Study , Genotype , High-Throughput Nucleotide Sequencing , Horses/physiology , Phenotype
19.
Nat Genet ; 50(4): 572-580, 2018 04.
Article in English | MEDLINE | ID: mdl-29632379

ABSTRACT

Individual risk of type 2 diabetes (T2D) is modified by perturbations to the mass, distribution and function of adipose tissue. To investigate the mechanisms underlying these associations, we explored the molecular, cellular and whole-body effects of T2D-associated alleles near KLF14. We show that KLF14 diabetes-risk alleles act in adipose tissue to reduce KLF14 expression and modulate, in trans, the expression of 385 genes. We demonstrate, in human cellular studies, that reduced KLF14 expression increases pre-adipocyte proliferation but disrupts lipogenesis, and in mice, that adipose tissue-specific deletion of Klf14 partially recapitulates the human phenotype of insulin resistance, dyslipidemia and T2D. We show that carriers of the KLF14 T2D risk allele shift body fat from gynoid stores to abdominal stores and display a marked increase in adipocyte cell size, and that these effects on fat distribution, and the T2D association, are female specific. The metabolic risk associated with variation at this imprinted locus depends on the sex both of the subject and of the parent from whom the risk allele derives.


Subject(s)
Adipocytes/pathology , Body Composition/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Sp Transcription Factors/genetics , Alleles , Animals , Body Fat Distribution , Cell Size , Enhancer Elements, Genetic , Female , Gene Expression , Genome-Wide Association Study , Genomic Imprinting , Humans , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Lipogenesis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Risk Factors , Sex Characteristics
20.
Cancer Prev Res (Phila) ; 11(7): 371-382, 2018 07.
Article in English | MEDLINE | ID: mdl-29545399

ABSTRACT

Specificity protein (Sp) transcription factors (TFs) such as Sp1 are critical for early development but their expression decreases with age and there is evidence that transformation of normal cells to cancer cells is associated with upregulation of Sp1, Sp3, and Sp4, which are highly expressed in cancer cells and tumors. Sp1 is a negative prognostic factor for pancreatic, colon, glioma, gastric, breast, prostate, and lung cancer patients. Functional studies also demonstrate that Sp TFs regulate genes responsible for cancer cell growth, survival, migration/invasion, inflammation and drug resistance, and Sp1, Sp3 and Sp4 are also nononcogene addiction (NOA) genes and important drug targets. The mechanisms of drug-induced downregulation of Sp TFs and pro-oncogenic Sp-regulated genes are complex and include ROS-dependent epigenetic pathways that initially decrease expression of the oncogene cMyc. Many compounds such as curcumin, aspirin, and metformin that are active in cancer prevention also exhibit chemotherapeutic activity and these compounds downregulate Sp TFs in cancer cell lines and tumors. The effects of these compounds on downregulation of Sp TFs in normal cells and the contribution of this response to their chemopreventive activity have not yet been determined. Cancer Prev Res; 11(7); 371-82. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Development/methods , Neoplasms/prevention & control , Sp Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/genetics , Disease Models, Animal , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Targeted Therapy/methods , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Prognosis , Sp Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...