Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.713
Filter
1.
J Nat Prod ; 87(4): 935-947, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38575516

ABSTRACT

We report on the use of nitric oxide-mediated transcriptional activation (NOMETA) as an innovative means to detect and access new classes of microbial natural products encoded within silent biosynthetic gene clusters. A small library of termite nest- and mangrove-derived fungi and actinomyces was subjected to cultivation profiling using a miniaturized 24-well format approach (MATRIX) in the presence and absence of nitric oxide, with the resulting metabolomes subjected to comparative chemical analysis using UPLC-DAD and GNPS molecular networking. This strategy prompted study of Talaromyces sp. CMB-TN6F and Coccidiodes sp. CMB-TN39F, leading to discovery of the triterpene glycoside pullenvalenes A-D (1-4), featuring an unprecedented triterpene carbon skeleton and rare 6-O-methyl-N-acetyl-d-glucosaminyl glycoside residues. Structure elucidation of 1-4 was achieved by a combination of detailed spectroscopic analysis, chemical degradation, derivatization and synthesis, and biosynthetic considerations.


Subject(s)
Aminoglycosides , Isoptera , Nitric Oxide , Triterpenes , Animals , Triterpenes/pharmacology , Triterpenes/chemistry , Triterpenes/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Molecular Structure , Isoptera/microbiology , Aminoglycosides/pharmacology , Australia , Transcriptional Activation/drug effects , Fungi/metabolism , Talaromyces/chemistry , Talaromyces/metabolism , Actinomyces/metabolism , Actinomyces/drug effects
2.
Angew Chem Int Ed Engl ; 63(21): e202400781, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38527936

ABSTRACT

Short amphipathic peptides are capable of binding to transcriptional coactivators, often targeting the same binding surfaces as native transcriptional activation domains. However, they do so with modest affinity and generally poor selectivity, limiting their utility as synthetic modulators. Here we show that incorporation of a medium-chain, branched fatty acid to the N-terminus of one such heptameric lipopeptidomimetic (LPPM-8) increases the affinity for the coactivator Med25 >20-fold (Ki >100 µM to 4 µM), rendering it an effective inhibitor of Med25 protein-protein interactions (PPIs). The lipid structure, the peptide sequence, and the C-terminal functionalization of the lipopeptidomimetic each influence the structural propensity of LPPM-8 and its effectiveness as an inhibitor. LPPM-8 engages Med25 through interaction with the H2 face of its activator interaction domain and in doing so stabilizes full-length protein in the cellular proteome. Further, genes regulated by Med25-activator PPIs are inhibited in a cell model of triple-negative breast cancer. Thus, LPPM-8 is a useful tool for studying Med25 and mediator complex biology and the results indicate that lipopeptidomimetics may be a robust source of inhibitors for activator-coactivator complexes.


Subject(s)
Mediator Complex , Humans , Mediator Complex/metabolism , Mediator Complex/chemistry , Protein Binding , Transcriptional Activation/drug effects , Peptides/chemistry , Peptides/pharmacology , Peptides/metabolism
3.
Phytomedicine ; 128: 155406, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38520834

ABSTRACT

BACKGROUND: Ischemic stroke (IS) is characterized as a detrimental cerebrovascular disease with high mortality and disability. Ferroptosis is a novel mechanism involved in neuronal death. There is a close connection between IS and ferroptosis, and inhibiting ferroptosis may provide an effective strategy for treating IS. Our previous investigations have discovered that kellerin, the active compound of Ferula sinkiangensis K. M. Shen, possesses the capability to shield against cerebral ischemia injury. PURPOSE: Our objective is to clarify the relationship between the neuroprotective properties of kellerin against IS and its ability to modulate ferroptosis, and investigate the underlying regulatory pathway. STUDY DESIGN: We investigated the impact and mechanism of kellerin in C57BL/6 mice underwent middle cerebral artery occlusion/reperfusion (MCAO/R) as well as SH-SY5Y cells exposed to oxygen-glucose deprivation/ re-oxygenation (OGD/R). METHODS: The roles of kellerin on neurological severity, cerebral infarction and edema were investigated in vivo. The regulatory impacts of kellerin on ferroptosis, mitochondrial damage and Akt/Nrf2 pathway were explored. Molecular docking combined with drug affinity responsive target stability assay (DARTS) and cellular thermal shift assay (CETSA) were performed to analyze the potential target proteins for kellerin. RESULTS: Kellerin protected against IS and inhibited ferroptosis in vivo. Meanwhile, kellerin improved the neuronal damage caused by OGD/R and suppressed ferroptosis by inhibiting the production of mitochondrial ROS in vitro. Further we found that kellerin directly interacted with Akt and enhanced its phosphorylation, leading to the increase of Nrf2 nuclear translocation and its downstream antioxidant genes expression. Moreover, kellerin's inhibitory effect on ferroptosis and mitochondrial ROS release was eliminated by inhibiting Akt/Nrf2 pathway. CONCLUSIONS: Our study firstly demonstrates that the neuroprotective properties of kellerin against IS are related to suppressing ferroptosis through inhibiting the production of mitochondrial ROS, in which its modulation on Akt-mediated transcriptional activation of Nrf2 plays an important role. This finding shed light on the potential mechanism that kellerin exerts therapeutic effects in IS.


Subject(s)
Ferroptosis , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Neuroprotective Agents , Proto-Oncogene Proteins c-akt , Animals , NF-E2-Related Factor 2/metabolism , Ferroptosis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Male , Mice , Humans , Neuroprotective Agents/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Brain Ischemia/drug therapy , Transcriptional Activation/drug effects , Reperfusion Injury/drug therapy , Cell Line, Tumor , Molecular Docking Simulation , Signal Transduction/drug effects
4.
Blood Cells Mol Dis ; 104: 102792, 2024 01.
Article in English | MEDLINE | ID: mdl-37633023

ABSTRACT

Sickle cell disease (SCD) is the most common ß-hemoglobinopathy caused by various mutations in the adult ß-globin gene resulting in sickle hemoglobin production, chronic hemolytic anemia, pain, and progressive organ damage. The best therapeutic strategies to manage the clinical symptoms of SCD is the induction of fetal hemoglobin (HbF) using chemical agents. At present, among the Food and Drug Administration-approved drugs to treat SCD, hydroxyurea is the only one proven to induce HbF protein synthesis, however, it is not effective in all people. Therefore, we evaluated the ability of the novel Bach1 inhibitor, HPP-D to induce HbF in KU812 cells and primary sickle erythroid progenitors. HPP-D increased HbF and decreased Bach1 protein levels in both cell types. Furthermore, chromatin immunoprecipitation assay showed reduced Bach1 and increased NRF2 binding to the γ-globin promoter antioxidant response elements. We also observed increased levels of the active histone marks H3K4Me1 and H3K4Me3 supporting an open chromatin configuration. In primary sickle erythroid progenitors, HPP-D increased γ-globin transcription and HbF positive cells and reduced sickled erythroid progenitors under hypoxia conditions. Collectively, our data demonstrate that HPP-D induces γ-globin gene transcription through Bach1 inhibition and enhanced NRF2 binding in the γ-globin promoter antioxidant response elements.


Subject(s)
Anemia, Sickle Cell , gamma-Globins , Humans , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , gamma-Globins/genetics , Hemoglobin, Sickle/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/therapeutic use , Transcriptional Activation/drug effects , Erythroid Cells/drug effects , Erythroid Cells/metabolism
5.
J Biol Chem ; 299(9): 105147, 2023 09.
Article in English | MEDLINE | ID: mdl-37567478

ABSTRACT

The vertebrate host's immune system and resident commensal bacteria deploy a range of highly reactive small molecules that provide a barrier against infections by microbial pathogens. Gut pathogens, such as Vibrio cholerae, sense and respond to these stressors by modulating the expression of exotoxins that are crucial for colonization. Here, we employ mass spectrometry-based profiling, metabolomics, expression assays, and biophysical approaches to show that transcriptional activation of the hemolysin gene hlyA in V. cholerae is regulated by intracellular forms of sulfur with sulfur-sulfur bonds, termed reactive sulfur species (RSS). We first present a comprehensive sequence similarity network analysis of the arsenic repressor superfamily of transcriptional regulators, where RSS and hydrogen peroxide sensors segregate into distinct clusters of sequences. We show that HlyU, transcriptional activator of hlyA in V. cholerae, belongs to the RSS-sensing cluster and readily reacts with organic persulfides, showing no reactivity or DNA dissociation following treatment with glutathione disulfide or hydrogen peroxide. Surprisingly, in V. cholerae cell cultures, both sulfide and peroxide treatment downregulate HlyU-dependent transcriptional activation of hlyA. However, RSS metabolite profiling shows that both sulfide and peroxide treatment raise the endogenous inorganic sulfide and disulfide levels to a similar extent, accounting for this crosstalk, and confirming that V. cholerae attenuates HlyU-mediated activation of hlyA in a specific response to intracellular RSS. These findings provide new evidence that gut pathogens may harness RSS-sensing as an evolutionary adaptation that allows them to overcome the gut inflammatory response by modulating the expression of exotoxins.


Subject(s)
Bacterial Proteins , Disulfides , Exotoxins , Gene Expression Regulation, Bacterial , Hemolysin Proteins , Intracellular Space , Sulfhydryl Compounds , Transcriptional Activation , Vibrio cholerae , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Exotoxins/genetics , Exotoxins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Transcriptional Activation/drug effects , Vibrio cholerae/drug effects , Vibrio cholerae/genetics , Vibrio cholerae/metabolism , Disulfides/metabolism , Disulfides/pharmacology , Sulfhydryl Compounds/metabolism , Sulfhydryl Compounds/pharmacology , Intracellular Space/metabolism , Mass Spectrometry , Metabolomics , Glutathione Disulfide/pharmacology , Gastrointestinal Microbiome/immunology
6.
Biosci Biotechnol Biochem ; 87(8): 907-915, 2023 Jul 24.
Article in English | MEDLINE | ID: mdl-37169920

ABSTRACT

We characterized the membrane vesicle fraction (RD-MV fraction) from bacterial strain RD055328, which is related to members of the genus Companilactobacillus and Lactiplantibacillus plantarum. RD-MVs and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) were detected in the RD-MV fraction. Immunoglobulin A (IgA) was produced by Peyer's patch cells following the addition of the RD-MV fraction. In the presence of the RD-MV fraction, RAW264 cells produced the pro-inflammatory cytokine IL-6. Recombinant GAPDH probably induced the production of IL-6 by RAW264 cells via superficial toll-like receptor 2 (TLR2) recognition. A confocal laser scanning microscopy image analysis indicated that RD-MVs and GAPDH were taken up by RAW264 cells. GAPDH wrapped around RAW264 cells. We suggest that GAPDH from strain RD055328 enhanced the production of IgA by acquired immune cells via the production of IL-6 by innate immune cells through TLR2 signal transduction.


Subject(s)
Bacterial Proteins , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating) , Lactobacillaceae , Signal Transduction , Toll-Like Receptor 2 , RAW 264.7 Cells , Signal Transduction/drug effects , Toll-Like Receptor 2/immunology , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Immunoglobulin A/immunology , Interleukin-6/immunology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/isolation & purification , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/pharmacology , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/isolation & purification , Adjuvants, Immunologic/pharmacology , Animals , Mice , Lactobacillaceae/classification , Lactobacillaceae/enzymology , Lactobacillaceae/genetics , Lactobacillaceae/isolation & purification , Bacterial Proteins/genetics , Bacterial Proteins/pharmacology , NF-kappa B/immunology , Transcriptional Activation/drug effects
7.
J Cell Mol Med ; 27(10): 1398-1409, 2023 05.
Article in English | MEDLINE | ID: mdl-37032511

ABSTRACT

Senescence of vascular endothelial cells is the major risk of vascular dysfunction and disease among elderly people. Parishin, which is a phenolic glucoside derived from Gastrodia elata, significantly prolonged yeast lifespan. However, the action of parishin in vascular ageing remains poorly understood. Here, we treated human coronary artery endothelial cells (HCAEC) and naturally aged mice by parishin. Parishin alleviated HCAEC senescence and general age-related features in vascular tissue in naturally aged mice. Network pharmacology approach was applied to determine the compound-target networks of parishin. Our analysis indicated that parishin had a strong binding affinity for Klotho. Expression of Klotho, a protein of age-related declines, was upregulated by parishin in serum and vascular tissue in naturally aged mice. Furthermore, FoxO1, on Klotho/FoxO1 signalling pathway, was increased in the parishin-intervened group, accompanied by the downregulated phosphorylated FoxO1. Taken together, parishin can increase Klotho expression to alleviate vascular endothelial cell senescence and vascular ageing.


Subject(s)
Aging , Glucosides , Klotho Proteins , Animals , Mice , Aging/blood , Aging/drug effects , Aging/genetics , Endothelial Cells , Klotho Proteins/blood , Klotho Proteins/metabolism , Transcriptional Activation/drug effects , Up-Regulation , Humans , Glucosides/pharmacology
8.
Chem Biol Interact ; 378: 110489, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37059213

ABSTRACT

We assessed the mechanism of human androgen receptor-mediated endocrine-disrupting effect by a triazole fungicide, metconazole in this study. The internationally validated stably transfected transactivation (STTA) in vitro assay, which was established for determination of a human androgen receptor (AR) agonist/antagonist by using 22Rv1/MMTV_GR-KO cell line, alongside an in vitro reporter-gene assay to confirm AR homodimerization was used. The STTA in vitro assay results showed that metconazole is a true AR antagonist. Furthermore, the results from the in vitro reporter-gene assay and western blotting showed that metconazole blocks the nuclear transfer of cytoplasmic AR proteins by suppressing the homodimerization of AR. These results suggest that metconazole can be considered to have an AR-mediated endocrine-disrupting effect. Additionally, the evidence from this study might help identify the endocrine-disrupting mechanism of triazole fungicides containing a phenyl ring.


Subject(s)
Androgen Receptor Antagonists , Endocrine Disruptors , Fungicides, Industrial , Protein Multimerization , Receptors, Androgen , Transcriptional Activation , Triazoles , Triazoles/chemistry , Triazoles/toxicity , Fungicides, Industrial/chemistry , Fungicides, Industrial/toxicity , Protein Multimerization/drug effects , Humans , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Endocrine Disruptors/chemistry , Endocrine Disruptors/pharmacology , Androgen Receptor Antagonists/chemistry , Androgen Receptor Antagonists/toxicity , Cell Line, Tumor , Transcriptional Activation/drug effects , Cytotoxins/chemistry , Cytotoxins/toxicity
9.
J Biol Chem ; 299(4): 104599, 2023 04.
Article in English | MEDLINE | ID: mdl-36907438

ABSTRACT

Immune cells adopt a variety of metabolic states to support their many biological functions, which include fighting pathogens, removing tissue debris, and tissue remodeling. One of the key mediators of these metabolic changes is the transcription factor hypoxia-inducible factor 1α (HIF-1α). Single-cell dynamics have been shown to be an important determinant of cell behavior; however, despite the importance of HIF-1α, little is known about its single-cell dynamics or their effect on metabolism. To address this knowledge gap, here we optimized a HIF-1α fluorescent reporter and applied it to study single-cell dynamics. First, we showed that single cells are likely able to differentiate multiple levels of prolyl hydroxylase inhibition, a marker of metabolic change, via HIF-1α activity. We then applied a physiological stimulus known to trigger metabolic change, interferon-γ, and observed heterogeneous, oscillatory HIF-1α responses in single cells. Finally, we input these dynamics into a mathematical model of HIF-1α-regulated metabolism and discovered a profound difference between cells exhibiting high versus low HIF-1α activation. Specifically, we found cells with high HIF-1α activation are able to meaningfully reduce flux through the tricarboxylic acid cycle and show a notable increase in the NAD+/NADH ratio compared with cells displaying low HIF-1α activation. Altogether, this work demonstrates an optimized reporter for studying HIF-1α in single cells and reveals previously unknown principles of HIF-1α activation.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Transcriptional Activation , Animals , Mice , Genes, Reporter/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Interferon-gamma/pharmacology , Mitochondria/metabolism , Models, Biological , Prolyl Hydroxylases/metabolism , RAW 264.7 Cells , Single-Cell Analysis/methods , Transcriptional Activation/drug effects
10.
Proc Natl Acad Sci U S A ; 119(28): e2206113119, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35867764

ABSTRACT

The Hippo signaling pathway acts as a brake on regeneration in many tissues. This cascade of kinases culminates in the phosphorylation of the transcriptional cofactors Yap and Taz, whose concentration in the nucleus consequently remains low. Various types of cellular signals can reduce phosphorylation, however, resulting in the accumulation of Yap and Taz in the nucleus and subsequently in mitosis. We earlier identified a small molecule, TRULI, that blocks the final kinases in the pathway, Lats1 and Lats2, and thus elicits proliferation of several cell types that are ordinarily postmitotic and aids regeneration in mammals. In the present study, we present the results of chemical modification of the original compound and demonstrate that a derivative, TDI-011536, is an effective blocker of Lats kinases in vitro at nanomolar concentrations. The compound fosters extensive proliferation in retinal organoids derived from human induced pluripotent stem cells. Intraperitoneal administration of the substance to mice suppresses Yap phosphorylation for several hours and induces transcriptional activation of Yap target genes in the heart, liver, and skin. Moreover, the compound initiates the proliferation of cardiomyocytes in adult mice following cardiac cryolesions. After further chemical refinement, related compounds might prove useful in protective and regenerative therapies.


Subject(s)
Protein Kinase Inhibitors , Protein Serine-Threonine Kinases , Regeneration , Animals , Cell Proliferation/drug effects , Heart/physiology , Humans , Induced Pluripotent Stem Cells , Liver Regeneration/drug effects , Liver Regeneration/genetics , Liver Regeneration/physiology , Mice , Organoids/physiology , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Regeneration/drug effects , Regeneration/genetics , Retina/physiology , Skin Physiological Phenomena/drug effects , Skin Physiological Phenomena/genetics , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , YAP-Signaling Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 119(31): e2201146119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35878041

ABSTRACT

Aberrant immune responses, including hyperresponsiveness to Toll-like receptor (TLR) ligands, underlie acute respiratory distress syndrome (ARDS). Type I interferons confer antiviral activities and could also regulate the inflammatory response, whereas little is known about their actions to resolve aberrant inflammation. Here we report that interferon-ß (IFN-ß) exerts partially overlapping, but also cooperative actions with aspirin-triggered 15-epi-lipoxin A4 (15-epi-LXA4) and 17-epi-resolvin D1 to counter TLR9-generated cues to regulate neutrophil apoptosis and phagocytosis in human neutrophils. In mice, TLR9 activation impairs bacterial clearance, prolongs Escherichia coli-evoked lung injury, and suppresses production of IFN-ß and the proresolving lipid mediators 15-epi-LXA4 and resolvin D1 (RvD1) in the lung. Neutralization of endogenous IFN-ß delays pulmonary clearance of E. coli and aggravates mucosal injury. Conversely, treatment of mice with IFN-ß accelerates clearance of bacteria, restores neutrophil phagocytosis, promotes neutrophil apoptosis and efferocytosis, and accelerates resolution of airway inflammation with concomitant increases in 15-epi-LXA4 and RvD1 production in the lungs. Pharmacological blockade of the lipoxin receptor ALX/FPR2 partially prevents IFN-ß-mediated resolution. These findings point to a pivotal role of IFN-ß in orchestrating timely resolution of neutrophil and TLR9 activation-driven airway inflammation and uncover an IFN-ß-initiated resolution program, activation of an ALX/FPR2-centered, proresolving lipids-mediated circuit, for ARDS.


Subject(s)
Interferon-beta , Lipoxins , Respiratory Distress Syndrome , Animals , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Escherichia coli , Escherichia coli Infections/immunology , Humans , Inflammation/drug therapy , Interferon-beta/immunology , Interferon-beta/pharmacology , Lipoxins/pharmacology , Mice , Receptors, Formyl Peptide/antagonists & inhibitors , Respiratory Distress Syndrome/drug therapy , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Transcriptional Activation/drug effects
12.
J Virol ; 96(15): e0037222, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35867565

ABSTRACT

Elimination of human immunodeficiency virus (HIV) reservoirs is a critical endpoint to eradicate HIV. One therapeutic intervention against latent HIV is "shock and kill." This strategy is based on the transcriptional activation of latent HIV with a latency-reversing agent (LRA) with the consequent killing of the reactivated cell by either the cytopathic effect of HIV or the immune system. We have previously found that the small molecule 3-hydroxy-1,2,3-benzotriazin-4(3H)-one (HODHBt) acts as an LRA by increasing signal transducer and activator of transcription (STAT) factor activation mediated by interleukin-15 (IL-15) in cells isolated from aviremic participants. The IL-15 superagonist N-803 is currently under clinical investigation to eliminate latent reservoirs. IL-15 and N-803 share similar mechanisms of action by promoting the activation of STATs and have shown some promise in preclinical models directed toward HIV eradication. In this work, we evaluated the ability of HODHBt to enhance IL-15 signaling in natural killer (NK) cells and the biological consequences associated with increased STAT activation in NK cell effector and memory-like functions. We showed that HODHBt increased IL-15-mediated STAT phosphorylation in NK cells, resulting in increases in the secretion of CXCL-10 and interferon gamma (IFN-γ) and the expression of cytotoxic proteins, including granzyme B, granzyme A, perforin, granulysin, FASL, and TRAIL. This increased cytotoxic profile results in increased cytotoxicity against HIV-infected cells and different tumor cell lines. HODHBt also improved the generation of cytokine-induced memory-like NK cells. Overall, our data demonstrate that enhancing the magnitude of IL-15 signaling with HODHBt favors NK cell cytotoxicity and memory-like generation, and thus, targeting this pathway could be further explored for HIV cure interventions. IMPORTANCE Several clinical trials targeting the HIV latent reservoir with LRAs have been completed. In spite of a lack of clinical benefit, they have been crucial to elucidate hurdles that "shock and kill" strategies have to overcome to promote an effective reduction of the latent reservoir to lead to a cure. These hurdles include low reactivation potential mediated by LRAs, the negative influence of some LRAs on the activity of natural killer and effector CD8 T cells, an increased resistance to apoptosis of latently infected cells, and an exhausted immune system due to chronic inflammation. To that end, finding therapeutic strategies that can overcome some of these challenges could improve the outcome of shock and kill strategies aimed at HIV eradication. Here, we show that the LRA HODHBt also improves IL-15-mediated NK cell effector and memory-like functions. As such, pharmacological enhancement of IL-15-mediated STAT activation can open new therapeutic avenues toward an HIV cure.


Subject(s)
HIV-1 , Immunologic Memory , Interleukin-15 , Killer Cells, Natural , STAT Transcription Factors , Triazines , Virus Latency , Humans , Cell Line, Tumor , Chemokine CXCL10 , Cytotoxicity Tests, Immunologic , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/growth & development , HIV-1/immunology , Immunologic Memory/drug effects , Interferon-gamma , Interleukin-15/immunology , Interleukin-15/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , STAT Transcription Factors/metabolism , Transcriptional Activation/drug effects , Triazines/pharmacology , Virus Activation/drug effects , Virus Latency/drug effects
13.
Oxid Med Cell Longev ; 2022: 7664290, 2022.
Article in English | MEDLINE | ID: mdl-35242277

ABSTRACT

Human cardiac fibroblasts (HCFs) play key roles in normal physiological functions and pathological processes in the heart. Our recent study has found that, in HCFs, sphingosine 1-phosphate (S1P) can upregulate the expression of cyclooxygenase-2 (COX-2) leading to prostaglandin E2 (PGE2) generation mediated by S1P receptors/PKCα/MAPKs cascade-dependent activation of NF-κB. Alternatively, G protein-coupled receptor- (GPCR-) mediated transactivation of receptor tyrosine kinases (RTKs) has been proved to induce inflammatory responses. However, whether GPCR-mediated transactivation of RTKs participated in the COX-2/PGE2 system induced by S1P is still unclear in HCFs. We hypothesize that GPCR-mediated transactivation of RTKs-dependent signaling cascade is involved in S1P-induced responses. This study is aimed at exploring the comprehensive mechanisms of S1P-promoted COX-2/PGE2 expression and apoptotic effects on HCFs. Here, we used pharmacological inhibitors and transfection with siRNA to evaluate whether matrix metalloprotease (MMP)2/9, heparin-binding- (HB-) epidermal growth factor (EGF), EGF receptor (EGFR), PI3K/Akt, MAPKs, and transcription factor AP-1 participated in the S1P-induced COX-2/PGE2 system determined by Western blotting, real-time polymerase chain reaction (RT-PCR), chromatin immunoprecipitation (ChIP), and promoter-reporter assays in HCFs. Our results showed that S1PR1/3 activated by S1P coupled to Gq- and Gi-mediated MMP9 activity to stimulate EGFR/PI3K/Akt/MAPKs/AP-1-dependent activity of transcription to upregulate COX-2 accompanied with PGE2 production, leading to stimulation of caspase-3 activity and apoptosis. Moreover, S1P-enhanced c-Jun bound to COX-2 promoters on its corresponding binding sites, which was attenuated by these inhibitors of protein kinases, determined by a ChIP assay. These results concluded that transactivation of MMP9/EGFR-mediated PI3K/Akt/MAPKs-dependent AP-1 activity was involved in the upregulation of the COX-2/PGE2 system induced by S1P, in turn leading to apoptosis in HCFs.


Subject(s)
Apoptosis/drug effects , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Fibroblasts/metabolism , Lysophospholipids/pharmacology , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 9/metabolism , Myocardium/cytology , Sphingosine/analogs & derivatives , Transcriptional Activation/drug effects , Up-Regulation/drug effects , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cyclooxygenase 2/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , MAP Kinase Signaling System/genetics , Matrix Metalloproteinase 9/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Sphingosine/pharmacology , Transcription Factor AP-1/metabolism , Transfection
14.
Cell Mol Life Sci ; 79(2): 121, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-35122536

ABSTRACT

Toll-like receptors (TLRs) recognise pathogen­associated molecular patterns, which allow the detection of microbial infection by host cells. Bacterial-derived toxin lipopolysaccharide activates TLR4 and leads to the activation of the Smad2 transcription factor. The phosphorylation of the Smad2 transcription factor is the result of the activation of the transforming growth factor-ß receptor 1 (TGFBR1). Therefore, we sought to investigate LPS via TLR4-mediated Smad2 carboxy terminal phosphorylation dependent on the transactivation of the TGFBR1. The in vitro model used human aortic vascular smooth muscle cells to assess the implications of TLR4 transactivation of the TGFBR1 in vascular pathophysiology. We show that LPS-mediated Smad2 carboxy terminal phosphorylation is inhibited in the presence of TGFBR1 inhibitor, SB431542. Treatment with MyD88 and TRIF pathway antagonists does not affect LPS-mediated phosphorylation of Smad2 carboxy terminal; however, LPS-mediated Smad2 phosphorylation was inhibited in the presence of MMP inhibitor, GM6001, and unaffected in the presence of ROCK inhibitor Y27632 or ROS/NOX inhibitor DPI. LPS via transactivation of the TGFBR1 stimulates PAI-1 mRNA expression. TLRs are first in line to respond to exogenous invading substances and endogenous molecules; our findings characterise a novel signalling pathway in the context of cell biology. Identifying TLR transactivation of the TGFBR1 may provide future insight into the detrimental implications of pathogens in pathophysiology.


Subject(s)
Lipopolysaccharides/pharmacology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Toll-Like Receptor 4/metabolism , Transcriptional Activation/drug effects , Benzamides/pharmacology , Cell Line , Dioxoles/pharmacology , Humans , Matrix Metalloproteinase 2/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myeloid Differentiation Factor 88/metabolism , Phosphorylation , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Smad2 Protein/metabolism , Up-Regulation/drug effects
15.
Int J Mol Sci ; 23(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35163379

ABSTRACT

Non-viral gene delivery has become a popular approach in tissue engineering, as it permits the transient delivery of a therapeutic gene, in order to stimulate tissue repair. However, the efficacy of non-viral delivery vectors remains an issue. Our lab has created gene-activated scaffolds by incorporating various non-viral delivery vectors, including the glycosaminoglycan-binding enhanced transduction (GET) peptide into collagen-based scaffolds with proven osteogenic potential. A modification to the GET peptide (FLR) by substitution of arginine residues with histidine (FLH) has been designed to enhance plasmid DNA (pDNA) delivery. In this study, we complexed pDNA with combinations of FLR and FLH peptides, termed GET* nanoparticles. We sought to enhance our gene-activated scaffold platform by incorporating GET* nanoparticles into collagen-nanohydroxyapatite scaffolds with proven osteogenic capacity. GET* N/P 8 was shown to be the most effective formulation for delivery to MSCs in 2D. Furthermore, GET* N/P 8 nanoparticles incorporated into collagen-nanohydroxyapatite (coll-nHA) scaffolds at a 1:1 ratio of collagen:nanohydroxyapatite was shown to be the optimal gene-activated scaffold. pDNA encoding stromal-derived factor 1α (pSDF-1α), an angiogenic chemokine which plays a role in BMP mediated differentiation of MSCs, was then delivered to MSCs using our optimised gene-activated scaffold platform, with the aim of significantly increasing angiogenesis as an important precursor to bone repair. The GET* N/P 8 coll-nHA scaffolds successfully delivered pSDF-1α to MSCs, resulting in a significant, sustained increase in SDF-1α protein production and an enhanced angiogenic effect, a key precursor in the early stages of bone repair.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Chemokine CXCL12/administration & dosage , Drug Delivery Systems , Neovascularization, Physiologic , Tissue Engineering , Tissue Scaffolds/chemistry , Transcriptional Activation , Animals , Biocompatible Materials/pharmacology , Chemokine CXCL12/pharmacology , Collagen/chemistry , DNA/chemistry , Durapatite/chemistry , Endothelial Progenitor Cells/metabolism , Glycosaminoglycans/chemistry , Nanoparticles , Neovascularization, Physiologic/drug effects , Plasmids/chemistry , Rats, Sprague-Dawley , Transcriptional Activation/drug effects , Transfection
16.
Cancer Lett ; 530: 170-180, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35077804

ABSTRACT

C/EBPß has recently emerged as a pro-leukemogenic transcription factor that cooperates with oncoprotein MYB to maintain proliferation and differentiation block of AML cells, making C/EBPß an interesting drug target for AML. Here we have studied the inhibitory potential and biological effects of a synthetic analog of the natural product helenalin, a known inhibitor of C/EBPß. The synthetic compound inhibits C/EBPß by covalent binding to cysteine residues in the transactivation domain, thereby causing up-regulation of differentiation-associated genes, cell death and reduced self-renewal potential of AML cells. Suppression of these effects by ectopic expression of C/EBPß or MYB and gene expression profiling validate C/EBPß as a relevant target of the helenalin-mimic and highlight its role as a pro-leukemogenic factor. Overall, our work demonstrates that the synthetic helenalin mimic acts as a covalent inhibitor of C/EBPß and identifies the cysteine residues in the transactivation domain of C/EBPß as ligandable sites. The helenalin mimic can be considered a potential "lead molecule" but needs further development towards more effective C/EBPß inhibitors before being used as a therapeutic agent.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/genetics , Leukemia, Myeloid, Acute/drug therapy , Sesquiterpenes, Guaiane/pharmacology , Transcriptional Activation/drug effects , 3T3 Cells , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HEK293 Cells , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Ligands , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , THP-1 Cells
17.
Eur J Clin Microbiol Infect Dis ; 41(4): 589-596, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35067799

ABSTRACT

Influenza A virus (IAV) H1N1 infection remains great challenge to public health and causes great burden over the world. Although there are anti-viral agents available, searching for effective agents to treat H1N1 infection is still in urgent because of the emergence of resistant strain. Protocatechuic acid (PCA) is a biological agent with multiple functions. In present study, we explored the effects of PCA on H1N1 infection. Mice infected with mouse adapted influenza strain A/Font Monmouth were administrated with PCA. The body weight change, mortality, lung index, viral titer, immune cell infiltration, and cytokine production in the lung were monitored. The activation of toll-like receptor 4 (TLR4) and nuclear factor kappa light chain enhancer of activated B cells (NF-κB) pathway was investigated. PCA treatment prevented H1N1 infection-induced mice body weight loss and death. PCA reduced the lung index, viral titer, infiltration of immune cells, and cytokine level in the lung, as well as suppressed H1N1-induced TLR4/NF-κB activation. PCA protects mice against H1N1 infection and could be a potential therapeutic agent to treat influenza.


Subject(s)
Hydroxybenzoates , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections , Animals , Disease Models, Animal , Hydroxybenzoates/pharmacology , Hydroxybenzoates/therapeutic use , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/prevention & control , Transcriptional Activation/drug effects
18.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34983841

ABSTRACT

Macrophages induce a number of inflammatory response genes in response to stimulation with microbial ligands. In response to endotoxin Lipid A, a gene-activation cascade of primary followed by secondary-response genes is induced. Epigenetic state is an important regulator of the kinetics, specificity, and mechanism of gene activation of these two classes. In particular, SWI/SNF chromatin-remodeling complexes are required for the induction of secondary-response genes, but not primary-response genes, which generally exhibit open chromatin. Here, we show that a recently discovered variant of the SWI/SNF complex, the noncanonical BAF complex (ncBAF), regulates secondary-response genes in the interferon (IFN) response pathway. Inhibition of bromodomain-containing protein 9 (BRD9), a subunit of the ncBAF complex, with BRD9 bromodomain inhibitors (BRD9i) or a degrader (dBRD9) led to reduction in a number of interferon-stimulated genes (ISGs) following stimulation with endotoxin lipid A. BRD9-dependent genes overlapped highly with a subset of genes differentially regulated by BET protein inhibition with JQ1 following endotoxin stimulation. We find that the BET protein BRD4 is cobound with BRD9 in unstimulated macrophages and corecruited upon stimulation to ISG promoters along with STAT1, STAT2, and IRF9, components of the ISGF3 complex activated downstream of IFN-alpha receptor stimulation. In the presence of BRD9i or dBRD9, STAT1-, STAT2-, and IRF9-binding is reduced, in some cases with reduced binding of BRD4. These results demonstrate a specific role for BRD9 and the ncBAF complex in ISG activation and identify an activity for BRD9 inhibitors and degraders in dampening endotoxin- and IFN-dependent gene expression.


Subject(s)
Cell Cycle Proteins/metabolism , Interferons/metabolism , Macrophage Activation/drug effects , Transcription Factors/metabolism , Antiviral Agents/pharmacology , Cell Cycle Proteins/genetics , Chromatin Assembly and Disassembly/drug effects , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-alpha/pharmacology , Interferons/genetics , Interferons/pharmacology , Promoter Regions, Genetic/drug effects , Protein Domains , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Transcription Factors/genetics , Transcriptional Activation/drug effects
19.
J Invest Dermatol ; 142(2): 390-401, 2022 02.
Article in English | MEDLINE | ID: mdl-34293351

ABSTRACT

Psoriasis is a chronic immune-mediated disease characterized by excessive proliferation of epidermal keratinocytes and increased immune cell infiltration to the skin. Although it is well-known that psoriasis pathogenesis is driven by aberrant production of proinflammatory cytokines, the mechanisms underlying the imbalance between proinflammatory and anti-inflammatory cytokine expression are incompletely understood. In this study, we report that the transcriptional coregulators CtBP1 and 2 can transactivate a common set of proinflammatory genes both in the skin of imiquimod-induced mouse psoriasis model and in human keratinocytes and macrophages stimulated by imiquimod. We find that mice overexpressing CtBP1 in epidermal keratinocytes display severe skin inflammation phenotypes with increased expression of T helper type 1 and T helper type 17 cytokines. We also find that the expression of CtBPs and CtBP-target genes is elevated both in human psoriatic lesions and in the mouse imiquimod psoriasis model. Moreover, we were able to show that topical treatment with a peptidic inhibitor of CtBP effectively suppresses the CtBP-regulated proinflammatory gene expression and thus attenuates psoriatic inflammation in the imiquimod mouse model. Together, our findings suggest to our knowledge previously unreported strategies for therapeutic modulation of the immune response in inflammatory skin diseases.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Psoriasis/drug therapy , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , HaCaT Cells , Humans , Imiquimod/immunology , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/pathology , Mice , Mice, Transgenic , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/pathology , Transcriptional Activation/drug effects , Transcriptional Activation/immunology
20.
Adv Sci (Weinh) ; 9(4): e2103669, 2022 02.
Article in English | MEDLINE | ID: mdl-34761556

ABSTRACT

Transcription activator RamA is linked to multidrug resistance of Klebsiella pneumoniae through controlling genes that encode efflux pumps (acrA) and porin-regulating antisense RNA (micF). In bacteria, σ70 , together with activators, controls the majority of genes by recruiting RNA polymerase (RNAP) to the promoter regions. RNAP and σ70 form a holoenzyme that recognizes -35 and -10 promoter DNA consensus sites. Many activators bind upstream from the holoenzyme and can be broadly divided into two classes. RamA acts as a class I activator on acrA and class II activator on micF, respectively. The authors present biochemical and structural data on RamA in complex with RNAP-σ70 at the two promoters and the data reveal the molecular basis for how RamA assembles and interacts with core RNAP and activates transcription that contributes to antibiotic resistance. Further, comparing with CAP/TAP complexes reveals common and activator-specific features in activator binding and uncovers distinct roles of the two C-terminal domains of RNAP α subunit.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Drug Resistance, Multiple, Bacterial/drug effects , Klebsiella pneumoniae/drug effects , Transcriptional Activation/drug effects , Anti-Bacterial Agents/metabolism , Bacterial Proteins/drug effects , Klebsiella pneumoniae/metabolism , Trans-Activators/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...