Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23.365
Filter
1.
Sci Rep ; 14(1): 22938, 2024 10 02.
Article in English | MEDLINE | ID: mdl-39358540

ABSTRACT

Activating transcription factor 4 (ATF4) plays a central role in the integrated stress response (ISR) and one overlapping branch of the unfolded protein response (UPR). We recently reported that the splicing inhibitor isoginkgetin (IGG) induced ATF4 protein along with several known ATF4-regulated transcripts in a response that resembled the ISR and UPR. However, the contribution of ATF4-dependent and -independent transcriptional responses to IGG exposure was not known. Here we used RNA-sequencing in HCT116 colon cancer cells and an isogenic subline lacking ATF4 to investigate the contribution of ATF4 to IGG-induced changes in gene expression. Approximately 85% of the IGG-responsive DEGs in HCT116 cells were also differentially expressed in response to the ER stressor thapsigargin (Tg) and these were enriched for genes associated with the UPR and ISR. Most of these were positively regulated by IGG with impaired responses in the ATF4-deficient cells. Nonetheless, there were DEGs that responded similarly in both cell lines. The ATF4-independent IGG-induced DEGs included several metal responsive transcripts encoding metallothionines and a zinc transporter. Taken together, the predominant IGG response was ATF4-dependent in these cells and resembled the UPR and ISR while a second less prominent response involved the ATF4-independent regulation of metal responsive mRNAs.


Subject(s)
Activating Transcription Factor 4 , Biflavonoids , Humans , HCT116 Cells , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Biflavonoids/pharmacology , Thapsigargin/pharmacology , Unfolded Protein Response/drug effects , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Transcription, Genetic/drug effects
2.
Sci Rep ; 14(1): 21165, 2024 09 10.
Article in English | MEDLINE | ID: mdl-39256449

ABSTRACT

Diabetes mellitus (DM) is a well-documented risk factor of intervertebral disc degeneration (IVDD). The current study was aimed to clarify the effects and mechanisms of NADH: ubiquinone oxidoreductase subunit A3 (NDUFA3) in human nucleus pulposus cells (HNPCs) exposed to high glucose. NDUFA3 was overexpressed in HNPCs via lenti-virus transduction, which were co-treated with high glucose and rotenone (a mitochondrial complex I inhibitor) for 48 h. Cell activities were assessed for cell viability, cell apoptosis, reactive oxygen species (ROS) production, mitochondrial membrane potential (MMP) ratio, oxygen consumption rate (OCR) and mitochondrial complexes I activities. High glucose decreased cell viability, increased apoptotic cells, increased ROS production, decreased MMP levels and OCR values in HNPCs in a dose-dependent manner. Rotenone co-treatment augmented the high glucose-induced injuries on cell viability, apoptosis, ROS production and mitochondrial function. NDUFA3 overexpression counteracted the high glucose-induced injuries in HNPCs. HDAC/H3K27ac mechanism was involved in regulating NDUFA3 transcription. NDUFA3 knockdown decreased cell viability and increased apoptotic cells, which were reversed by ROS scavenger N-acetylcysteine. HDAC/H3K27ac-mediated transcription of NDUFA3 protects HNPCs against high glucose-induced injuries through suppressing cell apoptosis, eliminating ROS, improving mitochondrial function and oxidative phosphorylation. This study sheds light on candidate therapeutic targets and deepens the understanding of molecular mechanisms behind DM-induced IVDD.


Subject(s)
Apoptosis , Electron Transport Complex I , Glucose , Histones , Mitochondria , Nucleus Pulposus , Humans , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Glucose/pharmacology , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histones/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Nucleus Pulposus/metabolism , Nucleus Pulposus/drug effects , Reactive Oxygen Species/metabolism , Rotenone/pharmacology , Transcription, Genetic/drug effects
3.
Ann Ist Super Sanita ; 60(3): 191-196, 2024.
Article in English | MEDLINE | ID: mdl-39269000

ABSTRACT

INTRODUCTION: We analyzed the expression of several genes implicated in the pathogenicity of Escherichia coli O157:H7, treating bacteria with Esc(1-21), a derivative of peptide esculentin-1 in combination with three essential oils obtained from plants from the Cympopogon genus. METHODS: We used the checkerboard assay to determine the antimicrobial activity of the combinations. We analyzed the expression of some genes implicated in the pathogenicity and quorum sensing system of E. coli O157:H7 by real-time RT-PCR technique. RESULTS: Treatment of the bacteria with the peptide combined with oils had an efficacious antimicrobial activity. The analysis of gene expression showed that all used combinations regulate positively the espAD and ler genes, located in the pathogenicity island, named the locus of enterocyte effacement. None of the combinations affects the quorum sensing genes: lsrABCFKR and qseBC. CONCLUSIONS: This study demonstrates that the use of essential oil/peptide combinations can be effective in fighting microbial infections.


Subject(s)
Cymbopogon , Escherichia coli O157 , Gene Expression Regulation, Bacterial , Oils, Volatile , Oils, Volatile/pharmacology , Escherichia coli O157/drug effects , Escherichia coli O157/genetics , Cymbopogon/chemistry , Gene Expression Regulation, Bacterial/drug effects , Transcription, Genetic/drug effects , Quorum Sensing/drug effects
4.
Pestic Biochem Physiol ; 204: 106102, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277424

ABSTRACT

While much attention has been devoted to understanding the transcriptional changes underlying resistance to insecticides, comparatively little is known about the transcriptional response of naive insects to agrochemicals. In this study, we analyze the transcriptomic response of an insecticide susceptible strain of Drosophila melanogaster to nine agrochemicals using a robust method that goes beyond classical replication standards. Our findings demonstrate that exposure to piperonyl butoxide (PBO), but not to eight other compounds, elicits a robust transcriptional response in a wild-type strain of Drosophila melanogaster. PBO exposure leads to the upregulation of a subset of Cyps, GSTs, UGTs and EcKls. This response is both time and concentration-dependent, suggesting that the degree of inhibition of P450 activity correlates with the magnitude of the transcriptional response. Furthermore, the upregulation of these enzymes is excluded from reproductive organs. Additionally, different sets of genes are regulated in the digestive/secretory tract and the carcass. Our results suggest that P450s play a role in metabolizing yet unidentified endogenous compounds and are involved in an as-yet-unknown physiological regulatory feedback loop.


Subject(s)
Cytochrome P-450 Enzyme System , Drosophila melanogaster , Piperonyl Butoxide , Animals , Piperonyl Butoxide/pharmacology , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Insecticides/toxicity , Insecticides/pharmacology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Transcription, Genetic/drug effects
5.
J Am Heart Assoc ; 13(18): e030941, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39248263

ABSTRACT

BACKGROUND: Mineralocorticoid receptor (MR) induces cardiac inflammation cooperatively with nuclear factor-κB and signal transducer and activator of transcription 3 (STAT3); MR blockers exert anti-inflammatory effects. However, the underlying mechanism remains unclear. We investigated the anti-inflammatory effect of esaxerenone, a novel MR blocker, in experimental myocardial infarction (MI) and its underlying mechanisms. METHODS AND RESULTS: Male C57BL/6J mice subjected to ligation of the left anterior descending artery were randomly assigned to either the vehicle or esaxerenone group. Esaxerenone was provided with a regular chow diet. The mice were euthanized at either 4 or 15 days after MI. Cardiac function, fibrosis, and inflammation were evaluated. Esaxerenone significantly improved cardiac function and attenuated cardiac fibrosis at 15 days after MI independently of its antihypertensive effect. Inflammatory cell infiltration, inflammatory-related gene expression, and elevated serum interleukin-6 levels at 4 days after MI were significantly attenuated by esaxerenone. In vitro experiments using mouse macrophage-like cell line RAW264.7 cells demonstrated that esaxerenone- and spironolactone-attenuated lipopolysaccharide-induced interleukin-6 expression without altering the posttranslational modification and nuclear translocation of p65 and STAT3. Immunoprecipitation assays revealed that MR interacted with both p65 and STAT3 and enhanced the p65-STAT3 interaction, leading to a subsequent increase in interleukin-6 promoter activity, which was reversed by esaxerenone. CONCLUSIONS: Esaxerenone ameliorated postinfarct remodeling in experimental MI through its anti-inflammatory properties exerted by modulating the transcriptional activity of the MR-p65-STAT3 complex. These results suggest that the MR-p65-STAT3 complex can be a novel therapeutic target for treating MI.


Subject(s)
Disease Models, Animal , Mice, Inbred C57BL , Mineralocorticoid Receptor Antagonists , Myocardial Infarction , Receptors, Mineralocorticoid , STAT3 Transcription Factor , Sulfones , Transcription Factor RelA , Animals , STAT3 Transcription Factor/metabolism , Male , Receptors, Mineralocorticoid/metabolism , Receptors, Mineralocorticoid/drug effects , Receptors, Mineralocorticoid/genetics , Mineralocorticoid Receptor Antagonists/pharmacology , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/drug therapy , Myocardial Infarction/prevention & control , Myocardial Infarction/genetics , Transcription Factor RelA/metabolism , RAW 264.7 Cells , Sulfones/pharmacology , Signal Transduction/drug effects , Fibrosis , Transcription, Genetic/drug effects , Myocardium/metabolism , Myocardium/pathology , Anti-Inflammatory Agents/pharmacology , Interleukin-6/metabolism , Interleukin-6/genetics , Pyrroles
6.
Antimicrob Agents Chemother ; 68(10): e0064324, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39230310

ABSTRACT

Human immunodeficiency virus-1 (HIV-1) encodes a transcriptional factor called Tat, which is critical for viral transcription. Tat-mediated transcription is highly ordered apart from the cellular manner; therefore, it is considered a target for developing anti-HIV-1 drugs. However, drugs targeting Tat-mediated viral transcription are not yet available. Our high-throughput screen of a compound library employing a dual-reporter assay identified a 1,3,4-oxadiazole scaffold against Tat and HIV-1 infection. Furthermore, a serial structure-activity relation (SAR) study performed with biological assays found 1,3,4-oxadiazole derivatives (9 and 13) containing indole and acetamide that exhibited potent inhibitory effects on HIV-1 infectivity, with half-maximal effective concentrations (EC50) of 0.17 (9) and 0.24 µM (13), respectively. The prominent derivatives specifically interfered with the viral transcriptional step without targeting other infection step(s) and efficiently inhibited the HIV-1 replication cycle in the T cell lines and peripheral blood mononuclear cells infected with HIV-1. Additionally, compared to the wild type, the compounds exhibited similar potency against anti-retroviral drug-resistant HIV-1 strains. In a series of mode-of-action studies, the compounds inhibited the ejection of histone H3 for facilitating viral transcription on the long-terminal repeat (LTR) promoter. Furthermore, SAHA (a histone deacetylase inhibitor) treatment abolished the compound potency, revealing that the compounds can possibly target Tat-regulated epigenetic modulation of LTR to inhibit viral transcription. Overall, our screening identified novel 1,3,4-oxadiazole compounds that inhibited HIV-1 Tat, and subsequent SAR-based optimization led to the derivatives 9 and 13 development that could be a promising scaffold for developing a new class of therapeutic agents for HIV-1 infection.


Subject(s)
Acetamides , Anti-HIV Agents , HIV-1 , Oxadiazoles , Transcription, Genetic , tat Gene Products, Human Immunodeficiency Virus , HIV-1/drug effects , HIV-1/genetics , Humans , Oxadiazoles/pharmacology , Oxadiazoles/chemistry , tat Gene Products, Human Immunodeficiency Virus/genetics , Anti-HIV Agents/pharmacology , Acetamides/pharmacology , Acetamides/chemistry , Transcription, Genetic/drug effects , Structure-Activity Relationship , Virus Replication/drug effects , Indoles/pharmacology , Indoles/chemistry , HIV Infections/drug therapy , HIV Infections/virology
7.
Psychopharmacology (Berl) ; 241(10): 1955-1981, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39254835

ABSTRACT

The persistent use of opioids leads to profound changes in neuroplasticity of the brain, contributing to the emergence and persistence of addiction. However, chronic opioid use disrupts the delicate balance of the reward system in the brain, leading to neuroadaptations that underlie addiction. Chronic cocaine usage leads to synchronized alterations in gene expression, causing modifications in the Nucleus Accumbens (NAc), a vital part of the reward system of the brain. These modifications assist in the development of maladaptive behaviors that resemble addiction. Neuroplasticity in the context of addiction involves changes in synaptic connectivity, neuronal morphology, and molecular signaling pathways. Drug-evoked neuroplasticity in opioid addiction and withdrawal represents a complicated interaction between environmental, genetic, and epigenetic factors. Identifying specific transcriptional and epigenetic targets that can be modulated to restore normal neuroplasticity without disrupting essential physiological processes is a critical consideration. The discussion in this article focuses on the transcriptional aspects of drug-evoked neuroplasticity, emphasizing the role of key transcription factors, including cAMP response element-binding protein (CREB), ΔFosB, NF-kB, Myocyte-enhancing factor 2 (MEF2), Methyl-CpG binding protein 2 (MeCP2), E2F3a, and FOXO3a. These factors regulate gene expression and lead to the neuroadaptive changes observed in addiction and withdrawal. Epigenetic regulation, which involves modifying gene accessibility by controlling these structures, has been identified as a critical component of addiction development. By unraveling these complex molecular processes, this study provides valuable insights that may pave the way for future therapeutic interventions targeting the mechanisms underlying addiction and withdrawal.


Subject(s)
Epigenesis, Genetic , Neuronal Plasticity , Opioid-Related Disorders , Substance Withdrawal Syndrome , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Humans , Animals , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/physiopathology , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/physiopathology , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Signal Transduction , Transcription, Genetic/drug effects
8.
Nat Commun ; 15(1): 6579, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097616

ABSTRACT

Bacteria often evolve antibiotic resistance through mutagenesis. However, the processes causing the mutagenesis have not been fully resolved. Here, we find that a broad range of ribosome-targeting antibiotics cause mutations through an underexplored pathway. Focusing on the clinically important aminoglycoside gentamicin, we find that the translation inhibitor causes genome-wide premature stalling of RNA polymerase (RNAP) in a loci-dependent manner. Further analysis shows that the stalling is caused by the disruption of transcription-translation coupling. Anti-intuitively, the stalled RNAPs subsequently induce lesions to the DNA via transcription-coupled repair. While most of the bacteria are killed by genotoxicity, a small subpopulation acquires mutations via SOS-induced mutagenesis. Given that these processes are triggered shortly after antibiotic addition, resistance rapidly emerges in the population. Our work reveals a mechanism of action of ribosomal antibiotics, illustrates the importance of dissecting the complex interplay between multiple molecular processes in understanding antibiotic efficacy, and suggests new strategies for countering the development of resistance.


Subject(s)
Anti-Bacterial Agents , DNA-Directed RNA Polymerases , Drug Resistance, Bacterial , Genomic Instability , Gentamicins , Ribosomes , Anti-Bacterial Agents/pharmacology , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , Ribosomes/metabolism , Ribosomes/drug effects , Gentamicins/pharmacology , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Mutation , Mutagenesis , Transcription, Genetic/drug effects , Protein Biosynthesis/drug effects
9.
Toxicol Appl Pharmacol ; 491: 117075, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39173720

ABSTRACT

Artesunate (ART) is a derivative of artemisinin and has anti-inflammatory, anti-tumor, and anti-angiogenic properties. Although ART has been implicated in osteoarthritis (OA), the mechanism needs to be further dissected. Here, we explored the effects of ART on the development of OA and the underlying mechanism using destabilization of the medial meniscus (DMM) surgical instability model. Mice with OA were developed using DMM and treated with ART. The pathological morphology of knee joint tissues was examined, and the degeneration of joint cartilage was assessed. Mouse knee chondrocytes were isolated and induced with IL-1ß, followed by ART treatment. ART alleviates OA in mice by elevating ubiquitin carboxyl-terminal hydrolase 7 (USP7) expression, and USP7 inhibitor (P22077) treatment mitigated the protective effects of ART on chondrocytes. We also showed that USP7 mediated the deubiquitination of forkhead box protein O1 (FoxO1), while FoxO1 alleviated chondrocyte injury. In addition, FoxO1 promoted metastasis-associated protein MTA1 (MTA1) transcription, and downregulation of MTA1 exacerbated chondrocyte injury. Our study identifies that USP7/FoxO1/MTA1 is a key signaling cascade in the treatment of ART on OA.


Subject(s)
Artesunate , Chondrocytes , Forkhead Box Protein O1 , Mice, Inbred C57BL , Osteoarthritis , Ubiquitin-Specific Peptidase 7 , Animals , Artesunate/pharmacology , Artesunate/therapeutic use , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin-Specific Peptidase 7/genetics , Mice , Male , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Trans-Activators/metabolism , Trans-Activators/genetics , Signal Transduction/drug effects , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription, Genetic/drug effects
10.
Nat Cardiovasc Res ; 3(3): 372-388, 2024 Mar.
Article in English | MEDLINE | ID: mdl-39183959

ABSTRACT

Targeting Meis1 and Hoxb13 transcriptional activity could be a viable therapeutic strategy for heart regeneration. In this study, we performd an in silico screening to identify FDA-approved drugs that can inhibit Meis1 and Hoxb13 transcriptional activity based on the resolved crystal structure of Meis1 and Hoxb13 bound to DNA. Paromomycin (Paro) and neomycin (Neo) induced proliferation of neonatal rat ventricular myocytes in vitro and displayed dose-dependent inhibition of Meis1 and Hoxb13 transcriptional activity by luciferase assay and disruption of DNA binding by electromobility shift assay. X-ray crystal structure revealed that both Paro and Neo bind to Meis1 near the Hoxb13-interacting domain. Administration of Paro-Neo combination in adult mice and in pigs after cardiac ischemia/reperfusion injury induced cardiomyocyte proliferation, improved left ventricular systolic function and decreased scar formation. Collectively, we identified FDA-approved drugs with therapeutic potential for induction of heart regeneration in mammals.


Subject(s)
Cell Proliferation , Homeodomain Proteins , Myeloid Ecotropic Viral Integration Site 1 Protein , Myocytes, Cardiac , Regeneration , Animals , Regeneration/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Cell Proliferation/drug effects , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Neomycin/pharmacology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Disease Models, Animal , Drug Approval , Mice , Ventricular Function, Left/drug effects , United States Food and Drug Administration , Rats , United States , Crystallography, X-Ray , Male , Mice, Inbred C57BL , Swine , Cells, Cultured , Transcription, Genetic/drug effects
11.
Pharmacol Res ; 208: 107377, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39209080

ABSTRACT

The bromodomain and extraterminal domain (BET) family proteins serve as primary readers of acetylated lysine residues and play crucial roles in cell proliferation and differentiation. Dysregulation of BET proteins has been implicated in tumorigenesis, making them important therapeutic targets. BET-bromodomain (BD) inhibitors and BET-targeting degraders have been developed to inhibit BET proteins. In this study, we found that the BET inhibitor MS645 exhibited superior antiproliferative activity than BET degraders including ARV771, AT1, MZ1 and dBET1 in triple-negative breast cancer (TNBC) cells. Treatment with MS645 led to the dissociation of BETs, MED1 and RNA polymerase II from the E2F1-3 promoter, resulting in the suppression of E2F1-3 transcription and subsequent inhibition of cell growth in TNBC. In contrast, while ARV771 displaced BET proteins from chromatin, it did not significantly alter E2F1-3 expression. Mechanistically, ARV771 induced BRD4 depletion at protein level, which markedly increased EGR1 expression. This elevation of EGR1 subsequently recruited septin 2 and septin 9 to E2F1-3 promoters, enhancing E2F1-3 transcription and promoting cell proliferation rate in vitro and in vivo. Our findings provide valuable insights into differential mechanisms of BET inhibition and highlight potential of developing BET-targeting molecules as therapeutic strategies for TNBC.


Subject(s)
Cell Proliferation , E2F1 Transcription Factor , Early Growth Response Protein 1 , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Humans , Cell Proliferation/drug effects , Animals , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/genetics , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Female , Cell Line, Tumor , E2F Transcription Factors/metabolism , E2F Transcription Factors/genetics , Antineoplastic Agents/pharmacology , Transcription, Genetic/drug effects , Mice, Nude , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Mice , Bromodomain Containing Proteins , Cell Cycle Proteins , E2F3 Transcription Factor
12.
EMBO Rep ; 25(9): 3944-3969, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39103676

ABSTRACT

The Hippo tumor suppressor pathway controls transcription by regulating nuclear abundance of YAP and TAZ, which activate transcription with the TEAD1-TEAD4 DNA-binding proteins. Recently, several small-molecule inhibitors of YAP and TEADs have been reported, with some entering clinical trials for different cancers with Hippo pathway deregulation, most notably, mesothelioma. Using genome-wide CRISPR/Cas9 screens we reveal that mutations in genes from the Hippo, MAPK, and JAK-STAT signaling pathways all modulate the response of mesothelioma cell lines to TEAD palmitoylation inhibitors. By exploring gene expression programs of mutant cells, we find that MAPK pathway hyperactivation confers resistance to TEAD inhibition by reinstating expression of a subset of YAP/TAZ target genes. Consistent with this, combined inhibition of TEAD and the MAPK kinase MEK, synergistically blocks proliferation of multiple mesothelioma and lung cancer cell lines and more potently reduces the growth of patient-derived lung cancer xenografts in vivo. Collectively, we reveal mechanisms by which cells can overcome small-molecule inhibition of TEAD palmitoylation and potential strategies to enhance the anti-tumor activity of emerging Hippo pathway targeted therapies.


Subject(s)
DNA-Binding Proteins , TEA Domain Transcription Factors , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Line, Tumor , Animals , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Hippo Signaling Pathway , Mice , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Cell Proliferation/drug effects , Lipoylation , Gene Expression Regulation, Neoplastic/drug effects , Transcription, Genetic/drug effects , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Small Molecule Libraries/pharmacology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mutation
13.
Br J Cancer ; 131(6): 1092-1105, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39117800

ABSTRACT

BACKGROUND: Cyclin-dependent kinase 9 (CDK9) stimulates oncogenic transcriptional pathways in cancer and CDK9 inhibitors have emerged as promising therapeutic candidates. METHODS: The activity of an orally bioavailable CDK9 inhibitor, CDKI-73, was evaluated in prostate cancer cell lines, a xenograft mouse model, and patient-derived tumor explants and organoids. Expression of CDK9 was evaluated in clinical specimens by mining public datasets and immunohistochemistry. Effects of CDKI-73 on prostate cancer cells were determined by cell-based assays, molecular profiling and transcriptomic/epigenomic approaches. RESULTS: CDKI-73 inhibited proliferation and enhanced cell death in diverse in vitro and in vivo models of androgen receptor (AR)-driven and AR-independent models. Mechanistically, CDKI-73-mediated inhibition of RNA polymerase II serine 2 phosphorylation resulted in reduced expression of BCL-2 anti-apoptotic factors and transcriptional defects. Transcriptomic and epigenomic approaches revealed that CDKI-73 suppressed signaling pathways regulated by AR, MYC, and BRD4, key drivers of dysregulated transcription in prostate cancer, and reprogrammed cancer-associated super-enhancers. These latter findings prompted the evaluation of CDKI-73 with the BRD4 inhibitor AZD5153, a combination that was synergistic in patient-derived organoids and in vivo. CONCLUSION: Our work demonstrates that CDK9 inhibition disrupts multiple oncogenic pathways and positions CDKI-73 as a promising therapeutic agent for prostate cancer, particularly aggressive, therapy-resistant subtypes.


Subject(s)
Cyclin-Dependent Kinase 9 , Epigenesis, Genetic , Prostatic Neoplasms , Male , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Humans , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Animals , Mice , Epigenesis, Genetic/drug effects , Cell Line, Tumor , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Transcription, Genetic/drug effects , Signal Transduction/drug effects
14.
JCI Insight ; 9(19)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39163135

ABSTRACT

Drugs that inhibit HIV transcription and/or reactivation of latent HIV have been proposed as a strategy to reduce HIV-associated immune activation or to achieve a functional cure, yet comparative studies are lacking. We evaluated 26 drugs, including drugs previously reported to inhibit HIV transcription (inhibitors of Tat-dependent HIV transcription, Rev, HSF-1/PTEF-b, HSP90, Jak/Stat, or SIRT1/Tat deacetylation) and other agents that were not tested before (inhibitors of PKC, NF-κB, SP-1, or histone acetyltransferase; NR2F1 agonists), elongation (inhibitors of CDK9/ PTEF-b), completion (inhibitors of PolyA-polymerase), or splicing (inhibitors of human splice factors). To investigate if those drugs would vary in their ability to affect different blocks to HIV transcription, we measured levels of initiated, elongated, midtranscribed, completed, and multiply spliced HIV RNA in PBMCs from antiretroviral therapy-suppressed individuals following ex vivo treatment with each drug and subsequent T cell activation. We identified new drugs that prevent HIV reactivation, including CDK and splicing inhibitors. While some drugs inhibited 1 or 2 steps, other drugs (CDK inhibitors, splicing inhibitors, tanespimycin, and triptolide) inhibited multiple stages of HIV transcription and blocked the production of supernatant viral RNA. These drugs and targets deserve further study in strategies aimed at reducing HIV-associated immune activation or achieving a functional cure.


Subject(s)
HIV Infections , HIV-1 , Virus Activation , Virus Latency , Humans , Virus Latency/drug effects , Virus Activation/drug effects , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Phenanthrenes/pharmacology , Phenanthrenes/therapeutic use , Diterpenes/pharmacology , Epoxy Compounds/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Transcription, Genetic/drug effects , Lymphocyte Activation/drug effects , RNA, Viral , Male , Pentacyclic Triterpenes
15.
Nat Commun ; 15(1): 6947, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138174

ABSTRACT

Fluxes in human copper levels recently garnered attention for roles in cellular signaling, including affecting levels of the signaling molecule cyclic adenosine monophosphate. We herein apply an unbiased temporal evaluation of the signaling and whole genome transcriptional activities modulated by copper level fluctuations to identify potential copper sensor proteins responsible for driving these activities. We find that fluctuations in physiologically relevant copper levels modulate EGFR signal transduction and activation of the transcription factor CREB. Both intracellular and extracellular assays support Cu1+ inhibition of the EGFR phosphatase PTPN2 (and potentially PTPN1)-via ligation to the PTPN2 active site cysteine side chain-as the underlying mechanism. We additionally show i) copper supplementation drives weak transcriptional repression of the copper importer CTR1 and ii) CREB activity is inversely correlated with CTR1 expression. In summary, our study reveals PTPN2 as a physiological copper sensor and defines a regulatory mechanism linking feedback control of copper stimulated EGFR/CREB signaling and CTR1 expression.


Subject(s)
Copper Transporter 1 , Copper , Cyclic AMP Response Element-Binding Protein , ErbB Receptors , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Signal Transduction , ErbB Receptors/metabolism , ErbB Receptors/genetics , Copper/metabolism , Humans , Cyclic AMP Response Element-Binding Protein/metabolism , Copper Transporter 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Transcription, Genetic/drug effects
16.
Free Radic Biol Med ; 223: 306-324, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39134162

ABSTRACT

The systemic inflammatory response syndrome (SIRS) represents a self-amplifying cascade of inflammatory reactions and pathophysiological states triggered by infectious or non-infectious factors. The identification of disease targets and differential proteins in the liver (the unique and important immune organ) of SIRS mice treated with the lead compound D1 was conducted using the Genecards database and proteomic analysis, respectively. Subsequently, NOTCH1 was identified as the potential hub target via an intersection analysis between the aforementioned differentially expressed proteins and disease targets. Based on our previous research on the structure-activity relationship, we designed and synthesized a series of SIRS-related derivatives, wherein butyl, halogen, and ester groups were incorporated into benzophenone, aiming at exploring the anti-inflammatory protective action from the perspective of macrophage polarization. Notably, these derivatives exhibited a direct binding capability to the O-glucosylation site (SER496) or its vicinities (such as SER492, VAL485) of NOTCH1 using docking, SPR, DARTS, and CETSA techniques. Mechanistically, derivative D6 exerted anti-inflammatory effects via the dual NOTCH pathway. Firstly, it could inhibit NOTCH1 nuclear transcriptional activity, attenuate the interaction between NICD and RBPJK, concurrently suppress NF-κB and NLRP3 inflammasome (NLRP3, ASC, and cleaved CASP1) activation, and promote NICD (NOTCH1 active fragments) ubiquitination metabolism (the nuclear transcriptional pathway). Secondly, it might possess the ability to increase PGC1α level, subsequently, enhance ATP and MMP levels, mitigate ROS production, increase mitochondrial numbers, and ameliorate mitochondrial inflammatory damage (the mitochondrial pathway). Importantly, the activator Jagged1 could effectively reverse the aforementioned effects, while the inhibitor DAPT exhibited a synergistic effect, suggesting that the nuclear transcriptional regulation and mitochondrial regulation were both in a NOTCH1-dependent manner. Subsequently, it effectively alleviated the inflammatory response and preserved organ function as evidenced by up-regulating M2-type macrophage-related anti-inflammatory cytokines (IL10, TGFß, CD206, and ARG1) and down-regulating M1-type macrophage-related pro-inflammatory cytokines (NO, IL6, IL18, iNOS, TNFα, CD86, and IL1ß). In a word, derivative D6 modulated macrophage polarization and effectively mitigated SIRS by targeting inhibition of the dual NOTCH pathway.


Subject(s)
Benzophenones , Mitochondria , Receptor, Notch1 , Signal Transduction , Systemic Inflammatory Response Syndrome , Animals , Benzophenones/pharmacology , Benzophenones/chemistry , Mice , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Signal Transduction/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Systemic Inflammatory Response Syndrome/drug therapy , Systemic Inflammatory Response Syndrome/metabolism , Systemic Inflammatory Response Syndrome/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Humans , Male , Molecular Docking Simulation , Inflammasomes/metabolism , Inflammasomes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Disease Models, Animal , RAW 264.7 Cells , Transcription, Genetic/drug effects , Structure-Activity Relationship
17.
BMC Genomics ; 25(1): 766, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39107687

ABSTRACT

BACKGROUND: Many common diseases exhibit uncontrolled mTOR signaling, prompting considerable interest in the therapeutic potential of mTOR inhibitors, such as rapamycin, to treat a range of conditions, including cancer, aging-related pathologies, and neurological disorders. Despite encouraging preclinical results, the success of mTOR interventions in the clinic has been limited by off-target side effects and dose-limiting toxicities. Improving clinical efficacy and mitigating side effects require a better understanding of the influence of key clinical factors, such as sex, tissue, and genomic background, on the outcomes of mTOR-targeting therapies. RESULTS: We assayed gene expression with and without rapamycin exposure across three distinct body parts (head, thorax, abdomen) of D. melanogaster flies, bearing either their native melanogaster mitochondrial genome or the mitochondrial genome from a related species, D. simulans. The fully factorial RNA-seq study design revealed a large number of genes that responded to the rapamycin treatment in a sex-dependent and tissue-dependent manner, and relatively few genes with the transcriptional response to rapamycin affected by the mitochondrial background. Reanalysis of an earlier study confirmed that mitochondria can have a temporal influence on rapamycin response. CONCLUSIONS: We found significant and wide-ranging effects of sex and body part, alongside a subtle, potentially time-dependent, influence of mitochondria on the transcriptional response to rapamycin. Our findings suggest a number of pathways that could be crucial for predicting potential side effects of mTOR inhibition in a particular sex or tissue. Further studies of the temporal response to rapamycin are necessary to elucidate the effects of the mitochondrial background on mTOR and its inhibition.


Subject(s)
Mitochondria , Sirolimus , Animals , Sirolimus/pharmacology , Female , Male , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/drug effects , Sex Factors , TOR Serine-Threonine Kinases/metabolism , Organ Specificity/genetics , Drosophila/genetics , Drosophila/drug effects , Transcription, Genetic/drug effects , Gene Expression Profiling
18.
Life Sci Alliance ; 7(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39089880

ABSTRACT

Ongoing viral transcription from the reservoir of HIV-1 infected long-lived memory CD4+ T cells presents a barrier to cure and associates with poorer health outcomes for people living with HIV, including chronic immune activation and inflammation. We previously reported that didehydro-cortistatin A (dCA), an HIV-1 Tat inhibitor, blocks HIV-1 transcription. Here, we examine the impact of dCA on host immune CD4+ T-cell transcriptional and epigenetic states. We performed a comprehensive analysis of genome-wide transcriptomic and DNA methylation profiles upon long-term dCA treatment of primary human memory CD4+ T cells. dCA prompted specific transcriptional and DNA methylation changes in cell cycle, histone, interferon-response, and T-cell lineage transcription factor genes, through inhibition of both HIV-1 and Mediator kinases. These alterations establish a tolerogenic Treg/Th2 phenotype, reducing viral gene expression and mitigating inflammation in primary CD4+ T cells during HIV-1 infection. In addition, dCA suppresses the expression of lineage-defining transcription factors for Th17 and Th1 cells, critical HIV-1 targets, and reservoirs. dCA's benefits thus extend beyond viral transcription inhibition, modulating the immune cell landscape to limit HIV-1 acquisition and inflammatory environment linked to HIV infection.


Subject(s)
CD4-Positive T-Lymphocytes , DNA Methylation , HIV Infections , HIV-1 , Heterocyclic Compounds, 4 or More Rings , Humans , HIV-1/drug effects , HIV-1/physiology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV Infections/genetics , DNA Methylation/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Transcription, Genetic/drug effects , Epigenesis, Genetic/drug effects , Th1 Cells/immunology , Th1 Cells/drug effects , Th1 Cells/metabolism , Isoquinolines
19.
PLoS One ; 19(8): e0304183, 2024.
Article in English | MEDLINE | ID: mdl-39088439

ABSTRACT

Expression of the androgen receptor is key to the response of cells and tissues to androgenic steroids, such as testosterone or dihydrotestosterone, as well as impacting the benefit of hormone-dependent therapies for endocrine diseases and hormone-dependent cancers. However, the mechanisms controlling androgen receptor expression are not fully understood, limiting our ability to effectively promote or inhibit androgenic signalling therapeutically. An autoregulatory loop has been described in which androgen receptor may repress its own expression in the presence of hormone, although the molecular mechanisms are not fully understood. In this work, we elucidate the mechanisms of autoregulation and demonstrate, for the first time, that a similar repression of the AR gene is facilitated by the progesterone receptor. We show that the progesterone receptor, like the androgen receptor binds to response elements within the AR gene to effect transcriptional repression in response to hormone treatment. Mechanistically, this repression involves hormone-dependent histone deacetylation within the AR 5'UTR region and looping between sequences in intron 2 and the transcription start site (TSS). This novel pathway controlling AR expression in response to hormone stimulation may have important implications for understanding cell or tissue selective receptor signalling.


Subject(s)
Gene Expression Regulation , Receptors, Androgen , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Humans , Gene Expression Regulation/drug effects , Receptors, Progesterone/metabolism , Receptors, Progesterone/genetics , 5' Untranslated Regions , Response Elements , Cell Line, Tumor , Acetylation , Transcription, Genetic/drug effects
20.
Cell Death Dis ; 15(8): 621, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39187513

ABSTRACT

Despite advancements in chemotherapy and the availability of novel therapies, the outcome of adult patients with B-cell acute lymphoblastic leukemia (B-ALL) remains unsatisfactory. Therefore, it is necessary to understand the molecular mechanisms underlying the progression of B-ALL. Brahma-related gene 1 (BRG1) is a poor prognostic factor for multiple cancers. Here, the expression of BRG1 was found to be higher in patients with B-ALL, irrespective of the molecular subtype, than in healthy individuals, and its overexpression was associated with a poor prognosis. Upregulation of BRG1 accelerated cell cycle progression into the S phase, resulting in increased cell proliferation, whereas its downregulation facilitated the apoptosis of B-ALL cells. Mechanistically, BRG1 occupies the transcriptional activation site of PPP2R1A, thereby inhibiting its expression and activating the PI3K/AKT signaling pathway to regulate the proto-oncogenes c-Myc and BCL-2. Consistently, silencing of BRG1 and administration of PFI-3 (a specific inhibitor targeting BRG1) significantly inhibited the progression of leukemia and effectively prolonged survival in cell-derived xenograft mouse models of B-ALL. Altogether, this study demonstrates that BRG1-induced overactivation of the PPP2R1A/PI3K/AKT signaling pathway plays an important role in promoting the progression of B-ALL. Therefore, targeting BRG1 represents a promising strategy for the treatment of B-ALL in adults.


Subject(s)
DNA Helicases , Disease Progression , Nuclear Proteins , Protein Phosphatase 2 , Transcription Factors , Animals , Female , Humans , Male , Mice , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Helicases/metabolism , DNA Helicases/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Transcription Factors/genetics , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL