Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.311
Filter
1.
Cell ; 182(4): 992-1008.e21, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32710817

ABSTRACT

Cellular heterogeneity confounds in situ assays of transcription factor (TF) binding. Single-cell RNA sequencing (scRNA-seq) deconvolves cell types from gene expression, but no technology links cell identity to TF binding sites (TFBS) in those cell types. We present self-reporting transposons (SRTs) and use them in single-cell calling cards (scCC), a novel assay for simultaneously measuring gene expression and mapping TFBS in single cells. The genomic locations of SRTs are recovered from mRNA, and SRTs deposited by exogenous, TF-transposase fusions can be used to map TFBS. We then present scCC, which map SRTs from scRNA-seq libraries, simultaneously identifying cell types and TFBS in those same cells. We benchmark multiple TFs with this technique. Next, we use scCC to discover BRD4-mediated cell-state transitions in K562 cells. Finally, we map BRD4 binding sites in the mouse cortex at single-cell resolution, establishing a new method for studying TF biology in situ.


Subject(s)
DNA Transposable Elements/genetics , Single-Cell Analysis/methods , Transcription Factors/metabolism , Animals , Binding Sites , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cerebral Cortex/metabolism , Chromatin Immunoprecipitation , Gene Expression , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Mice , Protein Binding , Sequence Analysis, RNA , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/genetics
2.
Cell ; 177(5): 1262-1279.e25, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31056284

ABSTRACT

Ferroptosis, a non-apoptotic form of programmed cell death, is triggered by oxidative stress in cancer, heat stress in plants, and hemorrhagic stroke. A homeostatic transcriptional response to ferroptotic stimuli is unknown. We show that neurons respond to ferroptotic stimuli by induction of selenoproteins, including antioxidant glutathione peroxidase 4 (GPX4). Pharmacological selenium (Se) augments GPX4 and other genes in this transcriptional program, the selenome, via coordinated activation of the transcription factors TFAP2c and Sp1 to protect neurons. Remarkably, a single dose of Se delivered into the brain drives antioxidant GPX4 expression, protects neurons, and improves behavior in a hemorrhagic stroke model. Altogether, we show that pharmacological Se supplementation effectively inhibits GPX4-dependent ferroptotic death as well as cell death induced by excitotoxicity or ER stress, which are GPX4 independent. Systemic administration of a brain-penetrant selenopeptide activates homeostatic transcription to inhibit cell death and improves function when delivered after hemorrhagic or ischemic stroke.


Subject(s)
Brain Ischemia , Cell-Penetrating Peptides/pharmacology , Ferroptosis/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Intracranial Hemorrhages , Neurons , Phospholipid Hydroperoxide Glutathione Peroxidase/biosynthesis , Selenium/pharmacology , Stroke , Transcription, Genetic/drug effects , Animals , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Humans , Intracranial Hemorrhages/drug therapy , Intracranial Hemorrhages/metabolism , Intracranial Hemorrhages/pathology , Male , Mice , Neurons/metabolism , Neurons/pathology , Sp1 Transcription Factor/metabolism , Stroke/drug therapy , Stroke/metabolism , Stroke/pathology , Transcription Factor AP-2/metabolism
3.
Mol Cell ; 82(17): 3135-3150.e9, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35914531

ABSTRACT

Alternative polyadenylation (APA) enhances gene regulatory potential by increasing the diversity of mRNA transcripts. 3' UTR shortening through APA correlates with enhanced cellular proliferation and is a widespread phenomenon in tumor cells. Here, we show that the ubiquitously expressed transcription factor Sp1 binds RNA in vivo and is a common repressor of distal poly(A) site usage. RNA sequencing identified 2,344 genes (36% of the total mapped mRNA transcripts) with lengthened 3' UTRs upon Sp1 depletion. Sp1 preferentially binds the 3' UTRs of such lengthened transcripts and inhibits cleavage at distal sites by interacting with the subunits of the core cleavage and polyadenylation (CPA) machinery. The 3' UTR lengths of Sp1 target genes in breast cancer patient RNA-seq data correlate with Sp1 expression levels, implicating Sp1-mediated APA regulation in modulating tumorigenic properties. Taken together, our findings provide insights into the mechanism for dynamic APA regulation by unraveling a previously unknown function of the DNA-binding transcription factor Sp1.


Subject(s)
Poly A , Polyadenylation , 3' Untranslated Regions , Humans , Poly A/metabolism , RNA, Messenger/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Zinc/metabolism
4.
Cell ; 158(3): 579-92, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083869

ABSTRACT

The p53 tumor suppressor coordinates a series of antiproliferative responses that restrict the expansion of malignant cells, and as a consequence, p53 is lost or mutated in the majority of human cancers. Here, we show that p53 restricts expression of the stem and progenitor-cell-associated protein nestin in an Sp1/3 transcription-factor-dependent manner and that Nestin is required for tumor initiation in vivo. Moreover, loss of p53 facilitates dedifferentiation of mature hepatocytes into nestin-positive progenitor-like cells, which are poised to differentiate into hepatocellular carcinomas (HCCs) or cholangiocarcinomas (CCs) in response to lineage-specific mutations that target Wnt and Notch signaling, respectively. Many human HCCs and CCs show elevated nestin expression, which correlates with p53 loss of function and is associated with decreased patient survival. Therefore, transcriptional repression of Nestin by p53 restricts cellular plasticity and tumorigenesis in liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Nestin/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic , Hepatocytes/metabolism , Humans , Liver Neoplasms/pathology , Mice , Prognosis , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
5.
Proc Natl Acad Sci U S A ; 121(29): e2401834121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38976739

ABSTRACT

Lung adenocarcinoma (LUAD) is the leading cause of cancer-related death worldwide, but the underlying molecular mechanisms remain largely unclear. The transcription factor (TF) specificity protein 1 (SP1) plays a crucial role in the development of various cancers, including LUAD. Recent studies have indicated that master TFs may form phase-separated macromolecular condensates to promote super-enhancer (SE) assembly and oncogene expression. In this study, we demonstrated that SP1 undergoes phase separation and that its zinc finger 3 in the DNA-binding domain is essential for this process. Through Cleavage Under Targets & Release Using Nuclease (CUT&RUN) using antibodies against SP1 and H3K27ac, we found a significant correlation between SP1 enrichment and SE elements, identified the regulator of the G protein signaling 20 (RGS20) gene as the most likely target regulated by SP1 through SE mechanisms, and verified this finding using different approaches. The oncogenic activity of SP1 relies on its phase separation ability and RGS20 gene activation, which can be abolished by glycogen synthase kinase J4 (GSK-J4), a demethylase inhibitor. Together, our findings provide evidence that SP1 regulates its target oncogene expression through phase separation and SE mechanisms, thereby promoting LUAD cell progression. This study also revealed an innovative target for LUAD therapies through intervening in SP1-mediated SE formation.


Subject(s)
Adenocarcinoma of Lung , Gene Expression Regulation, Neoplastic , Lung Neoplasms , RGS Proteins , Sp1 Transcription Factor , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Humans , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , RGS Proteins/metabolism , RGS Proteins/genetics , Cell Line, Tumor , Animals , Enhancer Elements, Genetic , Disease Progression , Mice , Phase Separation
6.
Development ; 150(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36762637

ABSTRACT

Members of the Sp family of transcription factors regulate gene expression via binding GC boxes within promoter regions. Unlike Sp1, which stimulates transcription, the closely related Sp3 can either repress or activate gene expression and is required for perinatal survival in mice. Here, we use RNA-seq and cellular phenotyping to show how Sp3 regulates murine fetal cell differentiation and proliferation. Homozygous Sp3-/- mice were smaller than wild-type and Sp+/- littermates, died soon after birth and had abnormal lung morphogenesis. RNA-seq of Sp3-/- fetal lung mesenchymal cells identified alterations in extracellular matrix production, developmental signaling pathways and myofibroblast/lipofibroblast differentiation. The lungs of Sp3-/- mice contained multiple structural defects, with abnormal endothelial cell morphology, lack of elastic fiber formation, and accumulation of lipid droplets within mesenchymal lipofibroblasts. Sp3-/- cells and mice also displayed cell cycle arrest, with accumulation in G0/G1 and reduced expression of numerous cell cycle regulators including Ccne1. These data detail the global impact of Sp3 on in vivo mouse gene expression and development.


Subject(s)
Embryonic Development , Transcription Factors , Animals , Mice , Cell Division , Lung , Promoter Regions, Genetic , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism
7.
J Biol Chem ; 300(2): 105605, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38159857

ABSTRACT

Prolidase (PEPD) is the only hydrolase that cleaves the dipeptides containing C-terminal proline or hydroxyproline-the rate-limiting step in collagen biosynthesis. However, the molecular regulation of prolidase expression remains largely unknown. In this study, we have identified overlapping binding sites for the transcription factors Krüppel-like factor 6 (KLF6) and Specificity protein 1 (Sp1) in the PEPD promoter and demonstrate that KLF6/Sp1 transcriptionally regulate prolidase expression. By cloning the PEPD promoter into a luciferase reporter and through site-directed deletion, we pinpointed the minimal sequences required for KLF6 and Sp1-mediated PEPD promoter-driven transcription. Interestingly, Sp1 inhibition abrogated KLF6-mediated PEPD promoter activity, suggesting that Sp1 is required for the basal expression of prolidase. We further studied the regulation of PEPD by KLF6 and Sp1 during transforming growth factor ß1 (TGF-ß1) signaling, since both KLF6 and Sp1 are key players in TGF-ß1 mediated collagen biosynthesis. Mouse and human fibroblasts exposed to TGF-ß1 resulted in the induction of PEPD transcription and prolidase expression. Inhibition of TGF-ß1 signaling abrogated PEPD promoter-driven transcriptional activity of KLF6 and Sp1. Knock-down of KLF6 as well as Sp1 inhibition also reduced prolidase expression. Chromatin immunoprecipitation assay supported direct binding of KLF6 and Sp1 to the PEPD promoter and this binding was enriched by TGF-ß1 treatment. Finally, immunofluorescence studies showed that KLF6 co-operates with Sp1 in the nucleus to activate prolidase expression and enhance collagen biosynthesis. Collectively, our results identify functional elements of the PEPD promoter for KLF6 and Sp1-mediated transcriptional activation and describe the molecular mechanism of prolidase expression.


Subject(s)
Dipeptidases , Kruppel-Like Factor 6 , Signal Transduction , Sp1 Transcription Factor , Animals , Humans , Mice , Collagen/metabolism , Kruppel-Like Factor 6/genetics , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
8.
J Virol ; 98(1): e0143623, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38084958

ABSTRACT

Bovine alphaherpesvirus 1 (BoHV-1) infections cause respiratory tract disorders and suppress immune responses, which can culminate in bacterial pneumonia. Following acute infection, BoHV-1 establishes lifelong latency in sensory neurons present in trigeminal ganglia (TG) and unknown cells in pharyngeal tonsil. Latently infected calves consistently reactivate from latency after an intravenous injection of the synthetic corticosteroid dexamethasone (DEX), which mimics the effects of stress. The immediate early transcription unit 1 (IEtu1) promoter drives expression of infected cell protein 0 (bICP0) and bICP4, two key viral transcriptional regulators. The IEtu1 promoter contains two functional glucocorticoid receptor (GR) response elements (GREs), and this promoter is transactivated by GR, DEX, and certain Krüppel transcription factors that interact with GC-rich motifs, including consensus specificity protein 1 (Sp1) binding sites. Based on these observations, we hypothesized that Sp1 stimulates productive infection and transactivates key BoHV-1 promoters. DEX treatment of latently infected calves increased the number of Sp1+ TG neurons and cells in pharyngeal tonsil indicating that Sp1 expression is induced by stress. Silencing Sp1 protein expression with siRNA or mithramycin A, a drug that preferentially binds GC-rich DNA, significantly reduced BoHV-1 replication. Moreover, BoHV-1 infection of permissive cells increased Sp1 steady-state protein levels. In transient transfection studies, GR and Sp1 cooperatively transactivate IEtu1 promoter activity unless both GREs are mutated. Co-immunoprecipitation studies revealed that GR and Sp1 interact in mouse neuroblastoma cells (Neuro-2A) suggesting this interaction stimulates IEtu1 promoter activity. Collectively, these studies suggested that the cellular transcription factor Sp1 enhances productive infection and stress-induced BoHV-1 reactivation from latency.IMPORTANCEFollowing acute infection, bovine alphaherpesvirus 1 (BoHV-1) establishes lifelong latency in sensory neurons in trigeminal ganglia (TG) and pharyngeal tonsil. The synthetic corticosteroid dexamethasone consistently induces BoHV-1 reactivation from latency. The number of TG neurons and cells in pharyngeal tonsil expressing the cellular transcription factor specificity protein 1 (Sp1) protein increases during early stages of dexamethasone-induced reactivation from latency. Silencing Sp1 expression impairs BoHV-1 replication in permissive cells. Interestingly, mithramycin A, a neuroprotective antibiotic that preferentially binds GC-rich DNA, impairs Sp1 functions and reduces BoHV-1 replication suggesting that it is a potential antiviral drug. The glucocorticoid receptor (GR) and Sp1 cooperatively transactivate the BoHV-1 immediate early transcript unit 1 (IEtu1) promoter, which drives expression of infected cell protein 0 (bICP0) and bICP4. Mithramycin A also reduced Sp1- and GR-mediated transactivation of the IEtu1 promoter. These studies revealed that GR and Sp1 trigger viral gene expression and replication following stressful stimuli.


Subject(s)
Herpesviridae Infections , Herpesvirus 1, Bovine , Receptors, Glucocorticoid , Sp1 Transcription Factor , Animals , Cattle , Mice , Adrenal Cortex Hormones/metabolism , Dexamethasone/pharmacology , DNA/metabolism , Herpesvirus 1, Bovine/physiology , Plicamycin/analogs & derivatives , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Transcription Factors/metabolism , Viral Proteins/metabolism , Sp1 Transcription Factor/metabolism
9.
J Virol ; 98(6): e0170523, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38742902

ABSTRACT

Long non-coding RNAs (lncRNAs) represent a new group of host factors involved in viral infection. Current study identified an intergenic lncRNA, LINC08148, as a proviral factor of Zika virus (ZIKV) and Dengue virus 2 (DENV2). Knockout (KO) or silencing of LINC08148 decreases the replication of ZIKV and DENV2. LINC08148 mainly acts at the endocytosis step of ZIKV but at a later stage of DENV2. RNA-seq analysis reveals that LINC08148 knockout downregulates the transcription levels of five endocytosis-related genes including AP2B1, CHMP4C, DNM1, FCHO1, and Src. Among them, loss of Src significantly decreases the uptake of ZIKV. Trans-complementation of Src in the LINC08148KO cells largely restores the caveola-mediated endocytosis of ZIKV, indicating that the proviral effect of LINC08148 is exerted through Src. Finally, LINC08148 upregulates the Src transcription through associating with its transcription factor SP1. This work establishes an essential role of LINC08148 in the ZIKV entry, underscoring a significance of lncRNAs in the viral infection. IMPORTANCE: Long non-coding RNAs (lncRNAs), like proteins, participate in viral infection. However, functions of most lncRNAs remain unknown. In this study, we performed a functional screen based on microarray data and identified a new proviral lncRNA, LINC08148. Then, we uncovered that LINC08148 is involved in the caveola-mediated endocytosis of ZIKV, rather than the classical clathrin-mediated endocytosis. Mechanistically, LINC08148 upregulates the transcription of Src, an initiator of caveola-mediated endocytosis, through binding to its transcription factor SP1. This study identifies a new lncRNA involved in the ZIKV infection, suggesting lncRNAs and cellular proteins are closely linked and cooperate to regulate viral infection.


Subject(s)
Endocytosis , RNA, Long Noncoding , Virus Internalization , Zika Virus Infection , Zika Virus , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , Zika Virus/genetics , Zika Virus/physiology , Humans , Zika Virus Infection/virology , Zika Virus Infection/metabolism , Zika Virus Infection/genetics , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Caveolae/metabolism , Animals , Virus Replication , Up-Regulation , Dengue Virus/physiology , Dengue Virus/genetics , Chlorocebus aethiops , HEK293 Cells , Vero Cells , src-Family Kinases/metabolism , src-Family Kinases/genetics
10.
J Virol ; 98(6): e0042324, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38771044

ABSTRACT

Bovine alphaherpesvirus 1 (BoHV-1) infection causes respiratory tract disorders and immune suppression and may induce bacterial pneumonia. BoHV-1 establishes lifelong latency in sensory neurons after acute infection. Reactivation from latency consistently occurs following stress or intravenous injection of the synthetic corticosteroid dexamethasone (DEX), which mimics stress. The immediate early transcription unit 1 (IEtu1) promoter drives expression of infected cell protein 0 (bICP0) and bICP4, two viral transcriptional regulators necessary for productive infection and reactivation from latency. The IEtu1 promoter contains two glucocorticoid receptor (GR) responsive elements (GREs) that are transactivated by activated GR. GC-rich motifs, including consensus binding sites for specificity protein 1 (Sp1), are in the IEtu1 promoter sequences. E2F family members bind a consensus sequence (TTTCCCGC) and certain specificity protein 1 (Sp1) sites. Consequently, we hypothesized that certain E2F family members activate IEtu1 promoter activity. DEX treatment of latently infected calves increased the number of E2F2+ TG neurons. GR and E2F2, but not E2F1, E2F3a, or E2F3b, cooperatively transactivate a 436-bp cis-regulatory module in the IEtu1 promoter that contains both GREs. A luciferase reporter construct containing a 222-bp fragment downstream of the GREs was transactivated by E2F2 unless two adjacent Sp1 binding sites were mutated. Chromatin immunoprecipitation studies revealed that E2F2 occupied IEtu1 promoter sequences when the BoHV-1 genome was transfected into mouse neuroblastoma (Neuro-2A) or monkey kidney (CV-1) cells. In summary, these findings revealed that GR and E2F2 cooperatively transactivate IEtu1 promoter activity, which is predicted to influence the early stages of BoHV-1 reactivation from latency. IMPORTANCE: Bovine alpha-herpesvirus 1 (BoHV-1) acute infection in cattle leads to establishment of latency in sensory neurons in the trigeminal ganglia (TG). A synthetic corticosteroid dexamethasone consistently initiates BoHV-1 reactivation in latently infected calves. The BoHV-1 immediate early transcription unit 1 (IEtu1) promoter regulates expression of infected cell protein 0 (bICP0) and bICP4, two viral transcriptional regulators. Hence, the IEtu1 promoter must be activated for the reactivation to occur. The number of TG neurons expressing E2F2, a transcription factor and cell cycle regulator, increased during early stages of reactivation from latency. The glucocorticoid receptor (GR) and E2F2, but not E2F1, E2F3a, or E2F3b, cooperatively transactivated a 436-bp cis-regulatory module (CRM) in the IEtu1 promoter that contains two GR responsive elements. Chromatin immunoprecipitation studies revealed that E2F2 occupies IEtu1 promoter sequences in cultured cells. GR and E2F2 mediate cooperative transactivation of IEtu1 promoter activity, which is predicted to stimulate viral replication following stressful stimuli.


Subject(s)
Cell Cycle , E2F2 Transcription Factor , Gene Expression Regulation, Viral , Herpesvirus 1, Bovine , Immediate-Early Proteins , Promoter Regions, Genetic , Receptors, Glucocorticoid , Transcriptional Activation , Animals , Cattle , Mice , Binding Sites , Cell Line , Dexamethasone/pharmacology , E2F2 Transcription Factor/metabolism , Gene Expression Regulation, Viral/drug effects , Gene Expression Regulation, Viral/genetics , Herpesviridae Infections/virology , Herpesviridae Infections/metabolism , Herpesviridae Infections/veterinary , Herpesviridae Infections/genetics , Herpesvirus 1, Bovine/genetics , Herpesvirus 1, Bovine/physiology , Immediate-Early Proteins/genetics , Neurons/virology , Receptors, Glucocorticoid/metabolism , Response Elements/genetics , Sp1 Transcription Factor/metabolism , Trans-Activators/metabolism , Trigeminal Ganglion/cytology , Trigeminal Ganglion/virology , Virus Activation , Virus Latency
11.
Exp Cell Res ; 438(2): 114050, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38663474

ABSTRACT

Myocardial infarction (MI) is a potentially fatal disease that causes a significant number of deaths worldwide. The strategy of increasing fatty acid oxidation in myocytes is considered a therapeutic avenue to accelerate metabolism to meet energy demands. We conducted the study aiming to investigate the effect of KN-93, which induces histone deacetylase (HDAC)4 shuttling to the nucleus, on fatty acid oxidation and the expression of related genes. A mouse model of myocardial infarction was induced by isoprenaline administration. Heart damage was assessed by the detection of cardiac injury markers. The level of fatty acid oxidation level was evaluated by testing the expression of related genes. Both immunofluorescence and immunoblotting in the cytosol or nucleus were utilized to observe the distribution of HDAC4. The interaction between HDAC4 and specificity protein (SP)1 was confirmed by co-immunoprecipitation. The acetylation level of SP1 was tested after KN-93 treatment and HDAC4 inhibitor. Oxygen consumption rate and immunoblotting experiments were used to determine whether the effect of KN-93 on increasing fatty acid oxidation is through HDAC4 and SP1. Administration of KN-93 significantly reduced cardiac injury in myocardial infarction and promoted fatty acid oxidation both in vitro and in vivo. KN-93 was shown to mediate nuclear translocation of HDAC4. HDAC4 was found to interact with SP1 and reduce SP1 acetylation. HDAC4 or SP1 inhibitors attenuated the effect of KN-93 on fatty acid oxidation. In conclusion, KN-93 promotes HDAC4 translocation to the nucleus, thereby potentially enhancing fatty acid oxidation by SP1.


Subject(s)
Cell Nucleus , Fatty Acids , Histone Deacetylases , Myocardial Infarction , Oxidation-Reduction , Animals , Humans , Male , Mice , Acetylation/drug effects , Active Transport, Cell Nucleus/drug effects , Cell Nucleus/metabolism , Fatty Acids/metabolism , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidation-Reduction/drug effects , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Benzylamines/pharmacology , Benzenesulfonamides/pharmacology
12.
Genomics ; 116(5): 110923, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39191354

ABSTRACT

BACKGROUND: Patients with lung adenocarcinoma (LUAD) generally have poor prognosis. The role of striatin-interacting protein 2 (STRIP2) in LUAD remain unclear. METHODS: Liquid chromatography-mass spectrometry analyses were used to screen the STRIP2-binding proteins and co-immunoprecipitation verified these interactions. A dual luciferase reporter assay explored the transcription factor activating STRIP2 transcription. Xenograft and lung metastasis models assessed STRIP2's role in tumor growth and metastasis in vivo. RESULTS: STRIP2 is highly expressed in LUAD tissues and is linked to poor prognosis. STRIP2 expression in LUAD cells significantly promoted cell proliferation, invasion, and migration in vitro and in vivo. Mechanistically, STRIP2 boosted the PI3K/AKT/mTOR/MYC cascades by binding AKT. In addition, specificity protein 1, potently activated STRIP2 transcription by binding to the STRIP2 promoter. Blocking STRIP2 reduces tumor growth and lung metastasis in xenograft models. CONCLUSIONS: Our study identifies STRIP2 is a key driver of LUAD progression and a potential therapeutic target.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Sp1 Transcription Factor , TOR Serine-Threonine Kinases , Humans , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Phosphatidylinositol 3-Kinases/metabolism , Mice , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Cell Line, Tumor , Mice, Nude , Cell Proliferation , A549 Cells , Cell Movement , Gene Expression Regulation, Neoplastic , Female , Male , Disease Progression
13.
J Mol Cell Cardiol ; 194: 59-69, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38880194

ABSTRACT

Obesity-induced cardiac dysfunction is growing at an alarming rate, showing a dramatic increase in global prevalence. Mitochondrial translocation of miR-181c in cardiomyocytes results in excessive reactive oxygen species (ROS) production during obesity. ROS causes Sp1, a transcription factor for MICU1, to be degraded via post-translational modification. The subsequent decrease in MICU1 expression causes mitochondrial Ca2+ accumulation, ultimately leading to a propensity for heart failure. Herein, we hypothesized that phosphorylation of Argonaute 2 (AGO2) at Ser 387 (in human) or Ser 388 (in mouse) inhibits the translocation of miR-181c into the mitochondria by increasing the cytoplasmic stability of the RNA-induced silencing complex (RISC). Initially, estrogen offers cardioprotection in pre-menopausal females against the consequences of mitochondrial miR-181c upregulation by driving the phosphorylation of AGO2. Neonatal mouse ventricular myocytes (NMVM) treated with insulin showed an increase in pAGO2 levels and a decrease in mitochondrial miR-181c expression by increasing the binding affinity of AGO2-GW182 in the RISC. Thus, insulin treatment prevented excessive ROS production and mitochondrial Ca2+ accumulation. In human cardiomyocytes, we overexpressed miR-181c to mimic pathological conditions, such as obesity/diabetes. Treatment with estradiol (E2) for 48 h significantly lowered miR-181c entry into the mitochondria through increased pAGO2 levels. E2 treatment also normalized Sp1 degradation and MICU1 transcription that normally occurs in response to miR-181c overexpression. We then investigated these findings using an in vivo model, with age-matched male, female and ovariectomized (OVX) female mice. Consistent with the E2 treatment, we show that female hearts express higher levels of pAGO2 and thus, exhibit higher association of AGO2-GW182 in cytoplasmic RISC. This results in lower expression of mitochondrial miR-181c in female hearts compared to male or OVX groups. Further, female hearts had fewer consequences of mitochondrial miR-181c expression, such as lower Sp1 degradation and significantly decreased MICU1 transcriptional regulation. Taken together, this study highlights a potential therapeutic target for conditions such as obesity and diabetes, where miR-181c is upregulated. NEW AND NOTEWORTHY: In this study, we show that the phosphorylation of Argonaute 2 (AGO2) stabilizes the RNA-induced silencing complex in the cytoplasm, preventing miR-181c entry into the mitochondria. Furthermore, we demonstrate that treatment with estradiol can inhibit the translocation of miR-181c into the mitochondria by phosphorylating AGO2. This ultimately eliminates the downstream consequences of miR-181c overexpression by mitigating excessive reactive oxygen species production and calcium entry into the mitochondria.


Subject(s)
Argonaute Proteins , MicroRNAs , Myocytes, Cardiac , Reactive Oxygen Species , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Female , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Male , Phosphorylation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Humans , Reactive Oxygen Species/metabolism , Mice , Mitochondria, Heart/metabolism , Calcium/metabolism , Sp1 Transcription Factor/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , RNA-Induced Silencing Complex/metabolism , Insulin/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Sex Characteristics
14.
Am J Physiol Cell Physiol ; 327(2): C387-C402, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38912734

ABSTRACT

RhoA and its effectors, the transcriptional coactivators myocardin-related transcription factor (MRTF) and serum response factor (SRF), control epithelial phenotype and are indispensable for profibrotic epithelial reprogramming during fibrogenesis. Context-dependent control of RhoA and fibrosis-associated changes in its regulators, however, remain incompletely characterized. We previously identified the guanine nucleotide exchange factor GEF-H1 as a central mediator of RhoA activation in renal tubular cells exposed to inflammatory or fibrotic stimuli. Here we found that GEF-H1 expression and phosphorylation were strongly elevated in two animal models of fibrosis. In the Unilateral Ureteral Obstruction mouse kidney fibrosis model, GEF-H1 was upregulated predominantly in the tubular compartment. GEF-H1 was also elevated and phosphorylated in a rat pulmonary artery banding (PAB) model of right ventricular fibrosis. Prolonged stimulation of LLC-PK1 tubular cells with tumor necrosis factor (TNF)-α or transforming growth factor (TGF)-ß1 increased GEF-H1 expression and activated a luciferase-coupled GEF-H1 promoter. Knockdown and overexpression studies revealed that these effects were mediated by RhoA, cytoskeleton remodeling, and MRTF, indicative of a positive feedback cycle. Indeed, silencing endogenous GEF-H1 attenuated activation of the GEF-H1 promoter. Of importance, inhibition of MRTF using CCG-1423 prevented GEF-H1 upregulation in both animal models. MRTF-dependent increase in GEF-H1 was prevented by inhibition of the transcription factor Sp1, and mutating putative Sp1 binding sites in the GEF-H1 promoter eliminated its MRTF-dependent activation. As the GEF-H1/RhoA axis is key for fibrogenesis, this novel MRTF/Sp1-dependent regulation of GEF-H1 abundance represents a potential target for reducing renal and cardiac fibrosis.NEW & NOTEWORTHY We show that expression of the RhoA regulator GEF-H1 is upregulated in tubular cells exposed to fibrogenic cytokines and in animal models of kidney and heart fibrosis. We identify a pathway wherein GEF-H1/RhoA-dependent MRTF activation through its noncanonical partner Sp1 upregulates GEF-H1. Our data reveal the existence of a positive feedback cycle that enhances Rho signaling through control of both GEF-H1 activation and expression. This feedback loop may play an important role in organ fibrosis.


Subject(s)
Fibrosis , Rho Guanine Nucleotide Exchange Factors , Sp1 Transcription Factor , Trans-Activators , rhoA GTP-Binding Protein , Animals , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , rhoA GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Mice , Rats , Feedback, Physiological , Male , Mice, Inbred C57BL , Humans , Signal Transduction , Swine , Phosphorylation , Disease Models, Animal , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology , Ureteral Obstruction/genetics , Rats, Sprague-Dawley , Cell Line , Transcription Factors
15.
J Biol Chem ; 299(11): 105351, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37838174

ABSTRACT

Breast cancer stem cells are mainly responsible for poor prognosis, especially in triple-negative breast cancer (TNBC). In a previous study, we demonstrated that ε-Sarcoglycan (SGCE), a type Ⅰ single-transmembrane protein, is a potential oncogene that promotes TNBC stemness by stabilizing EGFR. Here, we further found that SGCE depletion reduces breast cancer stem cells, partially through inhibiting the transcription of FGF-BP1, a secreted oncoprotein. Mechanistically, we demonstrate that SGCE could interact with the specific protein 1 transcription factor and translocate into the nucleus, which leads to an increase in the transcription of FGF-BP1, and the secreted FBF-BP1 activates FGF-FGFR signaling to promote cancer cell stemness. The novel SGCE-Sp1-FGF-BP1 axis provides novel potential candidate diagnostic markers and therapeutic targets for TNBC.


Subject(s)
Neoplastic Stem Cells , Sarcoglycans , Sp1 Transcription Factor , Triple Negative Breast Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , Neoplastic Stem Cells/metabolism , Sarcoglycans/metabolism , Signal Transduction , Sp1 Transcription Factor/metabolism , Triple Negative Breast Neoplasms/metabolism , Intercellular Signaling Peptides and Proteins/metabolism
16.
Am J Hum Genet ; 108(5): 903-918, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33909993

ABSTRACT

Macular degeneration (MD) is characterized by the progressive deterioration of the macula and represents one of the most prevalent causes of blindness worldwide. Abnormal intracellular accumulation of lipid droplets and pericellular deposits of lipid-rich material in the retinal pigment epithelium (RPE) called drusen are clinical hallmarks of different forms of MD including Doyne honeycomb retinal dystrophy (DHRD) and age-related MD (AMD). However, the appropriate molecular therapeutic target underlying these disorder phenotypes remains elusive. Here, we address this knowledge gap by comparing the proteomic profiles of induced pluripotent stem cell (iPSC)-derived RPEs (iRPE) from individuals with DHRD and their isogenic controls. Our analysis and follow-up studies elucidated the mechanism of lipid accumulation in DHRD iRPE cells. Specifically, we detected significant downregulation of carboxylesterase 1 (CES1), an enzyme that converts cholesteryl ester to free cholesterol, an indispensable process in cholesterol export. CES1 knockdown or overexpression of EFEMP1R345W, a variant of EGF-containing fibulin extracellular matrix protein 1 that is associated with DHRD and attenuated cholesterol efflux and led to lipid droplet accumulation. In iRPE cells, we also found that EFEMP1R345W has a hyper-inhibitory effect on epidermal growth factor receptor (EGFR) signaling when compared to EFEMP1WT and may suppress CES1 expression via the downregulation of transcription factor SP1. Taken together, these results highlight the homeostatic role of cholesterol efflux in iRPE cells and identify CES1 as a mediator of cholesterol efflux in MD.


Subject(s)
Cholesterol/metabolism , Macular Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , Adolescent , Adult , Carboxylic Ester Hydrolases/genetics , Cell Differentiation/genetics , Cytokines/metabolism , ErbB Receptors/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Inflammation/metabolism , Lipid Metabolism , Macular Degeneration/pathology , Middle Aged , Optic Disk Drusen/congenital , Optic Disk Drusen/metabolism , Proteomics , Proto-Oncogene Proteins c-akt/metabolism , Retinal Pigment Epithelium/pathology , Signal Transduction , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Unfolded Protein Response
17.
Mol Carcinog ; 63(6): 1174-1187, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38501385

ABSTRACT

Sorbin and SH3 domain-containing 2 (SORBS2) is an RNA-binding protein and has been implicated in the development of some cancers. However, its role in bladder cancer (BC) is yet to be established. The expression of SORBS2 in BC tissues was determined from the Gene Expression Omnibus and Gene Expression Profiling Interactive Analysis databases and collected paired tumor/normal samples. The effects of SORBS2 on BC cells were detected by CCK-8, colony formation, Transwell, dual-luciferase, RNA immunoprecipitation, chromatin immunoprecipitation, and DNA pull-down assays. In vivo, BC cell growth and metastasis were studied by a xenograft subcutaneous model and a tail-vein metastasis model. The results showed that SORBS2 expression was significantly decreased in BC tissues and cells. SORBS2 overexpression inhibited cell proliferation, migration, invasion, and epithelial-mesenchymal transition in vitro and tumor growth and metastasis in vivo, while silencing SORBS2 produced the opposite effect. Mechanistically, we found that SORBS2 enhanced the stability of tissue factor pathway inhibitor (TFPI) mRNA via direct binding to its 3' UTR. Restoration of TFPI expression reversed SORBS2 knockdown-induced malignant phenotypes of BC cells. In addition, SORBS2 expression was negatively regulated by the transcription factor specificity protein 1 (SP1). Conversely, SORBS2 can be transcriptionally regulated by SP1 and inhibit BC cell growth and metastasis via stabilization of TFPI mRNA, indicating SORBS2 may be a promising therapeutic target for BC.


Subject(s)
Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , RNA-Binding Proteins , Sp1 Transcription Factor , Urinary Bladder Neoplasms , Animals , Female , Humans , Male , Mice , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Mice, Inbred BALB C , Mice, Nude , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Xenograft Model Antitumor Assays , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism
18.
J Virol ; 97(10): e0111523, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37796122

ABSTRACT

IMPORTANCE: Of the flaviviruses, only CSFV and bovine viral diarrhea virus express Npro as the non-structural protein which is not essential for viral replication but functions to dampen host innate immunity. We have deciphered a novel mechanism with which CSFV uses to evade the host antiviral immunity by the N-terminal domain of its Npro to facilitate proteasomal degradation of Sp1 with subsequent reduction of HDAC1 and ISG15 expression. This is distinct from earlier findings involving Npro-mediated IRF3 degradation via the C-terminal domain. This study provides insights for further studies on how HDAC1 plays its role in antiviral immunity, and if and how other viral proteins, such as the core protein of CSFV, the nucleocapsid protein of porcine epidemic diarrhea virus, or even other coronaviruses, exert antiviral immune responses via the Sp1-HDAC1 axis. Such research may lead to a deeper understanding of viral immune evasion strategies as part of their pathogenetic mechanisms.


Subject(s)
Classical Swine Fever Virus , Classical Swine Fever , Endopeptidases , Histone Deacetylase 1 , Immunity, Innate , Proteasome Endopeptidase Complex , Sp1 Transcription Factor , Viral Proteins , Animals , Classical Swine Fever/immunology , Classical Swine Fever/metabolism , Classical Swine Fever/virology , Classical Swine Fever Virus/enzymology , Classical Swine Fever Virus/immunology , Classical Swine Fever Virus/metabolism , Classical Swine Fever Virus/pathogenicity , Endopeptidases/chemistry , Endopeptidases/metabolism , Histone Deacetylase 1/biosynthesis , Histone Deacetylase 1/metabolism , Interferon Regulatory Factor-3 , Nucleocapsid Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Sp1 Transcription Factor/metabolism , Swine/virology , Viral Core Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Ubiquitins/metabolism , Cytokines/metabolism , Porcine epidemic diarrhea virus/immunology , Porcine epidemic diarrhea virus/metabolism , Protein Domains
19.
Reproduction ; 168(5)2024 11 01.
Article in English | MEDLINE | ID: mdl-39133156

ABSTRACT

In brief: Cordycepin (COR), a compound derived from Cordyceps, is recognized as an adenosine analog with numerous beneficial effects on human health. However, its impact on steroidogenic acute regulatory protein (STAR) expression in ovarian granulosa cells is not well understood. This study demonstrates that COR downregulates STAR expression by reducing the expression of the SP1 transcription factor. Abstract: Cordycepin (COR), a pure compound of Cordyceps, is known as an adenosine analog that exerts many beneficial effects on human health. The steroidogenesis mediated by ovarian granulosa cells is pivotal in maintaining normal female reproductive function. The steroidogenic acute regulatory protein (STAR) regulates the rate-limiting step in steroidogenesis. COR has been shown to stimulate STAR expression in mouse Leydig cells, the steroidogenic cells in the testes. However, the effect of COR on STAR expression in ovarian granulosa cells remains undetermined. In the present study, we show that treatment with COR downregulates STAR expression in a steroidogenic human granulosa-like tumor cell line, KGN, and primary culture of human granulosa-lutein (hGL) cells obtained from patients undergoing in vitro fertilization. We used specific adenosine receptor (AR) antagonists, and our results reveal that the inhibitory effect of COR on STAR expression is mediated by AR-A1, AR-A2A, and AR-A3. In both KGN and primary hGL cells, COR activates ERK1/2 and AKT signaling pathways, but only activation of ERK1/2 is required for the COR-induced downregulation of STAR expression. In addition, our results demonstrate that COR downregulates STAR expression by reducing the expression of the SP1 transcription factor. These results provide a better understanding of the biological function of COR on STAR expression in the ovary, which may lead to the development of alternative therapeutic approaches for female reproductive disorders.


Subject(s)
Deoxyadenosines , Granulosa Cells , Luteal Cells , Phosphoproteins , Sp1 Transcription Factor , Female , Humans , Phosphoproteins/metabolism , Phosphoproteins/genetics , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Deoxyadenosines/pharmacology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Luteal Cells/drug effects , Luteal Cells/metabolism , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A1/genetics , Cell Line, Tumor , Signal Transduction/drug effects
20.
Exp Dermatol ; 33(6): e15112, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38840385

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC) ranks as the second most prevalent skin tumour (excluding melanoma). However, the molecular mechanisms driving cSCC progression remain elusive. This study aimed to investigate GBP1 expression in cSCC and elucidate its potential molecular mechanisms underlying cSCC development. GBP1 expression was assessed across public databases, cell lines and tissue samples. Various assays, including clone formation, CCK8 and EdU were employed to evaluate cell proliferation, while wound healing and transwell assays determined cell migration and invasion. Subcutaneous tumour assays were conducted to assess in vivo tumour proliferation, and molecular mechanisms were explored through western blotting, immunofluorescence and immunoprecipitation. Results identified GBP1 as an oncogene in cSCC, with elevated expression in both tumour tissues and cells, strongly correlating with tumour stage and grade. In vitro and in vivo investigations revealed that increased GBP1 expression significantly enhanced cSCC cell proliferation, migration and invasion. Mechanistically, GBP1 interaction with SP1 promoted STAT3 activation, contributing to malignant behaviours. In conclusion, the study highlights the crucial role of the GBP1/SP1/STAT3 signalling axis in regulating tumour progression in cSCC. These findings provide valuable insights into the molecular mechanisms of cSCC development and offer potential therapeutic targets for interventions against cSCC.


Subject(s)
Carcinoma, Squamous Cell , Cell Movement , Cell Proliferation , GTP-Binding Proteins , Neoplasm Invasiveness , STAT3 Transcription Factor , Skin Neoplasms , Sp1 Transcription Factor , STAT3 Transcription Factor/metabolism , Humans , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/genetics , Sp1 Transcription Factor/metabolism , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , Cell Line, Tumor , Animals , Mice , Signal Transduction , Female , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL