Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
J Med Chem ; 67(4): 2812-2836, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38329974

ABSTRACT

Homo sapiens caseinolytic protease P (HsClpP) activation is a promising strategy for colon cancer treatment. In this study, CCG1423 was identified as a selective activator of HsClpP. After optimization, NCA029 emerged as the most potent compound, with an EC50 of 0.2 µM against HsClpP. Molecular dynamics revealed that the affinity of NCA029 for the YYW aromatic network is crucial for its selectivity toward HsClpP. Furthermore, NCA029 displayed favorable pharmacokinetics and safety profiles and significantly inhibited tumor growth in HCT116 xenografts, resulting in 83.6% tumor inhibition. Mechanistically, NCA029 targeted HsClpP, inducing mitochondrial dysfunction and activating the ATF3-dependent integrated stress response, ultimately causing cell death in colorectal adenocarcinoma. These findings highlight NCA029 as an effective HsClpP activator with potential for colon cancer therapy.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Colorectal Neoplasms , Humans , Colorectal Neoplasms/drug therapy , Colonic Neoplasms/pathology , Peptide Hydrolases , Apoptosis , Cell Line, Tumor , Activating Transcription Factor 3/pharmacology , Activating Transcription Factor 3/physiology
2.
J Clin Invest ; 131(22)2021 11 15.
Article in English | MEDLINE | ID: mdl-34779419

ABSTRACT

Genetic variants near the TRIB1 gene are highly significantly associated with plasma lipid traits and coronary artery disease. While TRIB1 is likely causal of these associations, the molecular mechanisms are not well understood. Here we sought to investigate how TRIB1 influences low density lipoprotein cholesterol (LDL-C) levels in mice. Hepatocyte-specific deletion of Trib1 (Trib1Δhep) in mice increased plasma cholesterol and apoB and slowed the catabolism of LDL-apoB due to decreased levels of LDL receptor (LDLR) mRNA and protein. Simultaneous deletion of the transcription factor CCAAT/enhancer-binding protein alpha (CEBPα) with TRIB1 eliminated the effects of TRIB1 on hepatic LDLR regulation and LDL catabolism. Using RNA-seq, we found that activating transcription factor 3 (Atf3) was highly upregulated in the livers of Trib1Δhep but not Trib1Δhep CebpaΔhep mice. ATF3 has been shown to directly bind to the CEBPα protein, and to repress the expression of LDLR by binding its promoter. Blunting the increase of ATF3 in Trib1Δhep mice reduced the levels of plasma cholesterol and partially attenuated the effects on LDLR. Based on these data, we conclude that deletion of Trib1 leads to a posttranslational increase in CEBPα, which increases ATF3 levels, thereby contributing to the downregulation of LDLR and increased plasma LDL-C.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/physiology , Hepatocytes/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Lipoproteins, LDL/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, LDL/analysis , Activating Transcription Factor 3/physiology , Animals , Apolipoproteins B/metabolism , Female , Humans , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/physiology
3.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34298975

ABSTRACT

Previously, we showed that chemotherapy paradoxically exacerbated cancer cell colonization at the secondary site in a manner dependent on Atf3, a stress-inducible gene, in the non-cancer host cells. Here, we present evidence that this phenotype is established at an early stage of colonization within days of cancer cell arrival. Using mouse breast cancer models, we showed that, in the wild-type (WT) lung, cyclophosphamide (CTX) increased the ability of the lung to retain cancer cells in the vascular bed. Although CTX did not change the WT lung to affect cancer cell extravasation or proliferation, it changed the lung macrophage to be pro-cancer, protecting cancer cells from death. This, combined with the initial increase in cell retention, resulted in higher lung colonization in CTX-treated than control-treated mice. In the Atf3 knockout (KO) lung, CTX also increased the ability of lung to retain cancer cells. However, the CTX-treated KO macrophage was highly cytotoxic to cancer cells, resulting in no increase in lung colonization-despite the initial increase in cell retention. In summary, the status of Atf3 dictates the dichotomous activity of macrophage: pro-cancer for CTX-treated WT macrophage but anti-cancer for the KO counterpart. This dichotomy provides a mechanistic explanation for CTX to exacerbate lung colonization in the WT but not Atf3 KO lung.


Subject(s)
Activating Transcription Factor 3/physiology , Cyclophosphamide/toxicity , Lung Neoplasms/secondary , Macrophages/physiology , Mammary Neoplasms, Experimental/genetics , Neoplasm Metastasis/physiopathology , Neoplasm Proteins/physiology , Stress, Physiological/genetics , Tumor-Associated Macrophages/physiology , Animals , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/genetics , Cell Line, Tumor , Cyclophosphamide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Genotype , Humans , Lung Neoplasms/metabolism , Macrophage Activation , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Neoadjuvant Therapy/adverse effects , Neoplasm Metastasis/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation/methods , Neoplastic Stem Cells/pathology , Transendothelial and Transepithelial Migration , Tumor Microenvironment , Tumor-Associated Macrophages/drug effects , Cathelicidins
4.
Nat Metab ; 3(1): 59-74, 2021 01.
Article in English | MEDLINE | ID: mdl-33462514

ABSTRACT

Activating transcription factor (ATF)3 is known to have an anti-inflammatory function, yet the role of hepatic ATF3 in lipoprotein metabolism or atherosclerosis remains unknown. Here we show that overexpression of human ATF3 in hepatocytes reduces the development of atherosclerosis in Western-diet-fed Ldlr-/- or Apoe-/- mice, whereas hepatocyte-specific ablation of Atf3 has the opposite effect. We further show that hepatic ATF3 expression is inhibited by hydrocortisone. Mechanistically, hepatocyte ATF3 enhances high-density lipoprotein (HDL) uptake, inhibits intestinal fat and cholesterol absorption and promotes macrophage reverse cholesterol transport by inducing scavenger receptor group B type 1 (SR-BI) and repressing cholesterol 12α-hydroxylase (CYP8B1) in the liver through its interaction with p53 and hepatocyte nuclear factor 4α, respectively. Our data demonstrate that hepatocyte ATF3 is a key regulator of HDL and bile acid metabolism and atherosclerosis.


Subject(s)
Activating Transcription Factor 3/physiology , Atherosclerosis/prevention & control , Bile Acids and Salts/metabolism , Hepatocytes/metabolism , Lipoproteins, HDL/metabolism , Animals , Apolipoproteins E/genetics , Cholesterol, Dietary/metabolism , Dietary Fats/metabolism , Gene Expression Regulation/drug effects , Hepatocyte Nuclear Factor 4/metabolism , Humans , Hydrocortisone/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/genetics , Scavenger Receptors, Class B/metabolism , Steroid 12-alpha-Hydroxylase/metabolism , Tumor Suppressor Protein p53/metabolism
5.
Hum Cell ; 34(1): 49-59, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32959354

ABSTRACT

ATF3 (activating transcription factor 3) is a member of the mammalian activation transcription factor/cAMP-responsive element-binding (CREB) family. It plays a role in inflammation and innate immunity, and suggests that ATF3 is associated with atherosclerosis. In our study, we analyzed datasets of atherosclerosis from the NCBI-GEO (Gene Expression Omnibus) database and found that expression levels of ATF3 were lower in macrophages from ruptured atherosclerotic plaques than from stable atherosclerotic plaques. Expression levels of ATF3 correlated with the stability of atherosclerotic plaques. KEGG analysis of different expression genes (DEGs) between ruptured and stable atherosclerotic plaques was performed by Metascape database. The PI3K-AKT pathway may be a potential pathway of the formation of ruptured atherosclerotic plaques. High-fat diet-induced atherosclerosis apoE-/- mice were divided into two groups: a model group and an ATF3 overexpression (OE)-group. Tests on atherosclerotic plaques in the aortic root suggested that absence of ATF3 and increase of macrophages may be risk factors for the formation of ruptured atherosclerotic plaques. We found decreased areas of lesions in aortic roots and branches of aortic arch, as well as increased lesional content of macrophages as well as TUNEL-positive areas. Consistent with these results, we found reduced degradation and incidence of elastic plate cracks accompanied by suppressed MMPs expression and transduction pathway protein PI3K/AKT activation. These data suggest that ATF3 is a signaling molecule that mediates the progression and stability of atherosclerotic plaques. ATF3 could be a potential new biomarker for the prognosis of atherosclerosis and may be a therapeutic target to reduce atherosclerosis.


Subject(s)
Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/physiology , Atherosclerosis/diagnosis , Atherosclerosis/genetics , Plaque, Atherosclerotic/genetics , Activating Transcription Factor 3/metabolism , Animals , Atherosclerosis/immunology , Atherosclerosis/therapy , Biomarkers , Gene Expression , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases/metabolism , Plaque, Atherosclerotic/etiology , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
6.
Hum Cell ; 34(1): 177-186, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32936424

ABSTRACT

The aim of this study was to investigate the genes associated with ferroptosis and the progression of hepatocellular carcinoma (HCC). The RNA sequencing data of erastin-induced ferroptosis in HCC cells were downloaded from the Sequence Read Archive database with accession number SRP119173. The microarray dataset GSE89377 of HCC progression was downloaded from the Gene Expression Omnibus database. The ferroptosis-related genes were screened by differential analysis and HCC progression-related genes were screened by cluster analysis using Mfuzz. Then, the genes associated with ferroptosis and HCC progression were screened by Venn analysis, followed by functional enrichment, protein-protein interaction (PPI) analysis, and transcription factor (TF) prediction. Finally, survival analysis was performed using data from the Cancer Genome Atlas database. A total of 33 upregulated and 52 downregulated genes associated with HCC progression and ferroptosis were obtained, and these genes were significantly involved in the negative regulation of ERK1 and ERK2 cascades; the NAD biosynthetic process; alanine, aspartate, and glutamate metabolism; and other pathways. The PPI network contained 52 genes and 78 interactions, of which, cell division cycle 20 (CDC20) and heat shock protein family B (small) member 1 (HSPB1) were hub genes found in higher degrees. Among the 85 genes associated with HCC progression and ferroptosis, two TFs (activating TF 3 (ATF3) and HLF) were predicted, with HSPB1 targeted by ATF3. In addition, 26 genes that were found to be significantly correlated with the overall survival of HCC patients were screened, including CDC20 and thyroid hormone receptor interactor 13. Several genes associated with HCC progression and ferroptosis were screened based on a comprehensive bioinformatics analysis. These genes played roles in HCC progression and ferroptosis via the negative regulation of the ERK1 and ERK2 cascades; the NAD biosynthetic process; and alanine, aspartate, and glutamate metabolism. ATF3 and HSPB1 played important roles in HCC progression and ferroptosis, with HSPB1 possibly regulated by ATF3.


Subject(s)
Activating Transcription Factor 3/physiology , Carcinoma, Hepatocellular/genetics , Ferroptosis/genetics , Gene Expression Regulation, Neoplastic/genetics , Genetic Association Studies , Heat-Shock Proteins/physiology , Liver Neoplasms/genetics , Molecular Chaperones/physiology , Alanine/metabolism , Disease Progression , Glutamic Acid/metabolism , Humans , Liver Neoplasms/mortality , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , NAD/biosynthesis , Survival Rate
7.
Nephrology (Carlton) ; 26(4): 358-368, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33295061

ABSTRACT

INTRODUCTION: Nicotiflorin is the main characteristic component of Nymphaea candida, which is a natural product that reportedly ameliorates acute injury of the liver and cerebral cortex, but the effect of nicotiflorin on acute kidney injury (AKI) remains unknown. This study aimed to investigate the effects of nicotiflorin on ischaemia/reperfusion (I/R) AKI and the associated mechanisms. METHODS: We performed both (a) in vivo experiments with C57BL/6 mice with bilateral renal pedicles clamped for 45 minutes and (b) in vitro experiments with human kidney epithelial cells (HK-2) exposed to hypoxia/reoxygenation to mimic I/R injury to study the role of nicotiflorin in AKI. RESULTS: In vivo, nicotiflorin administration exerted protective effects on renal injury, as demonstrated by reductions in the levels of caspase3 and Bad (P < .05), the upregulation of Bcl-2 expression (P < .05) and improved renal histologic changes, which suggested that nicotiflorin can alleviate I/R injury and cell apoptosis. In vitro, nicotiflorin at a concentration of 75 µg/mL protected cells from hypoxia, which further confirmed that nicotiflorin exerts beneficial effects on hypoxia/reoxygenation. Through computational molecular docking, we found that activating transcription factor 3 (ATF3) exhibits a robust interaction with nicotiflorin with a simulated binding energy of -9.2°. We verified the interaction of nicotiflorin with ATF3 in HK-2 cells, and found that nicotiflorin reduced the apoptosis of HK-2 through ATF3. CONCLUSION: Based on the above-described results, nicotiflorin appears to have a beneficial impact on deteriorated renal function, as demonstrated using an experimental I/R model. The underlying mechanisms of nicotiflorin might inhibit HK-2 cell apoptosis through ATF3.


Subject(s)
Activating Transcription Factor 3/drug effects , Activating Transcription Factor 3/physiology , Apoptosis/drug effects , Flavonoids/pharmacology , Flavonoids/therapeutic use , Kidney/blood supply , Phenols/pharmacology , Phenols/therapeutic use , Reperfusion Injury/drug therapy , Animals , Mice , Mice, Inbred C57BL
8.
Cell Death Dis ; 11(8): 695, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32826874

ABSTRACT

Endothelial to mesenchymal transition (EndMT) is an important pathological change in many diseases. Semaphorin7A (Sema7A) has been reported to regulate nerve and vessel homeostasis, but its role in EndMT remains unclear. Here we investigate the effect of Sema7A on EndMT and the underlying mechanism. Sema7A-overexpressed human umbilical vein endothelial cells (Sema7A-HUVECs) were generated and showed lower levels of endothelial cell markers and higher levels of mesenchymal cell markers indicating the occurrence of EndMT. RNA-sequencing analysis showed a total of 1168 upregulated genes and 886 downregulated genes. Among them, most of the molecules associated with EndMT were upregulated in Sema7A-HUVECs. Mechanistically, Sema7A-HUVECs showed a higher TGF-ß2 expression and activated TGF-ß/Smad Signaling. Importantly, Sema7A overexpression upregulated activating transcription factor 3 (ATF3) that was found to selectively bind the promotor region of TGF-ß2, but not TGF-ß1, promoting TGF-ß2 transcription, which was further confirmed by ATF3-siRNA knockdown approach. Blocking ß1 integrin, a known Sema7A receptor, alleviated the expression of ATF3, TGF-ß2, and EndMT in Sema7A-overexpressed HUVECs, implying a role of ß1 integrin/ATF3/TGF-ß2 axis in mediating Sema7A-induced EndMT. Using Sema7A-deficient mice and the partial carotid artery ligation (PCL) model, we showed that Sema7A deletion attenuated EndMT induced by blood flow disturbance in vivo. In conclusion, Sema7A promotes TGF-ß2 secretion by upregulating transcription factor ATF3 in a ß1 integrin-dependent manner, and thus facilitates EndMT through TGF/Smad signaling, implying Sema7A as a potential therapeutic target for EndMT-related vascular diseases.


Subject(s)
Activating Transcription Factor 3/metabolism , Antigens, CD/genetics , Epithelial-Mesenchymal Transition/physiology , Semaphorins/genetics , Activating Transcription Factor 3/physiology , Animals , Antigens, CD/metabolism , Cell Movement/drug effects , Cells, Cultured , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Integrin beta1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Semaphorins/metabolism , Signal Transduction/drug effects , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/metabolism
9.
Cancer Lett ; 486: 38-45, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32428661

ABSTRACT

Atypical teratoid/rhabdoid tumor (AT/RT) is the most malignant tumor of the central nervous system that generally occurs in young children. Despite the use of intensive multimodal therapy for AT/RT, the prognosis is still poor. The brain tumor initiating cells in AT/RT cells has been suggested as one of the challenges in AT/RT treatment. These cells have high expression of aldehyde dehydrogenase (ALDH). We investigated the combination effect of the ALDH inhibitor, disulfiram and cisplatin in the treatment of AT/RT cells. Isobologram analysis revealed that the combination therapy synergistically increases AT/RT cell death. The enzyme activity of ALDH AT/RT cells was effectively reduced by the combination therapy. We proposed that the synergistic augmentation occurs, at least partially through an increase in cleaved Poly (ADP-ribose) polymerase (PARP)-dependent apoptosis mediated by activating transcription factor 3 (ATF3). In the AT/RT mouse model, the combination therapy decreased tumor volume and prolonged survival. Immunofluorescence assay in mouse brain tissues were consistent with the expression of ATF3 and cleaved PARP. Our study demonstrates enhanced anti-cancer effect of combination therapy of disulfiram and cisplatin. This combination might provide a viable therapeutic strategy for AT/RT patients.


Subject(s)
Brain Neoplasms/drug therapy , Cisplatin/pharmacology , Disulfiram/pharmacology , Rhabdoid Tumor/drug therapy , Teratoma/drug therapy , Activating Transcription Factor 3/physiology , Aldehyde Dehydrogenase/metabolism , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cisplatin/administration & dosage , Disulfiram/administration & dosage , Drug Therapy, Combination , Female , Humans , Mice , Mice, Inbred BALB C , Rhabdoid Tumor/pathology , Teratoma/pathology
10.
Invest Ophthalmol Vis Sci ; 61(2): 31, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32084268

ABSTRACT

Purpose: To investigate the possible role of activating transcription factor 3 (ATF3) in retinal ganglion cell (RGC) neuroprotection and optic nerve regeneration after optic nerve crush (ONC). Methods: Overexpression of proteins of interest (ATF3, phosphatase and tensin homolog [PTEN], placental alkaline phosphatase, green fluorescent protein) in the retina was achieved by intravitreal injections of recombinant adenovirus-associated viruses (rAAVs) expressing corresponding proteins. The number of RGCs and αRGCs was evaluated by immunostaining retinal sections and whole-mount retinas with antibodies against RNA binding protein with multiple splicing (RBPMS) and osteopontin, respectively. Axonal regeneration was assessed via fluorophore-coupled cholera toxin subunit B labeling. RGC function was evaluated by recording positive scotopic threshold response. Results: The level of ATF3 is preferentially elevated in osteopontin+/RBPMS+ αRGCs following ONC. Overexpression of ATF3 by intravitreal injection of rAAV 2 weeks before ONC promoted RBPMS+ RGC survival and preserved RGC function as assessed by positive scotopic threshold response recordings 2 weeks after ONC. However, overexpression of ATF3 and simultaneous downregulation of PTEN, a negative regulator of the mTOR pathway, combined with ONC, only moderately promoted short distance RGC axon regeneration (200 µm from the lesion site) but did not provide additional RGC neuroprotection compared with PTEN downregulation alone. Conclusions: These results reveal a neuroprotective effect of ATF3 in the retina following injury and identify ATF3 as a promising agent for potential treatments of optic neuropathies.


Subject(s)
Activating Transcription Factor 3/physiology , Neuroprotection/physiology , Optic Nerve Injuries/physiopathology , Retinal Ganglion Cells/physiology , Activating Transcription Factor 3/metabolism , Animals , Axons/pathology , Mice , Mice, Inbred C57BL , Nerve Crush , Nerve Regeneration/physiology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/pathology
11.
Eur Rev Med Pharmacol Sci ; 23(23): 10300-10312, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31841185

ABSTRACT

OBJECTIVE: Circular RNAs (circRNAs) have been demonstrated to involve in the development of various cancers. This study aimed to investigate the functions of circ_0001742 on regulating tongue squamous cell carcinoma (TSCC) development and the underlying mechanisms. PATIENTS AND METHODS: The expression of circ_0001742, miR-431-5p and activating transcription factor 3 (ATF3) mRNA was detected by quantitative real-time polymerase chain reaction (qRT-PCR). The protein levels of epithelial-mesenchymal transition (EMT)-related proteins and ATF3 were measured by Western blot analysis. 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and flow cytometry assay were used to evaluate cell proliferation and apoptosis. Besides, Cell migration and invasion were assessed by transwell assay. The relationships between circ_0001742 and miR-431-5p, miR-431-5p and ATF3 were predicted by online software and confirmed by dual-luciferase reporter assay, RNA immunoprecipitation (RIP), and pull-down assay. RESULTS: The expression of circ_0001742 was upregulated in TSCC tissues and cells. Knockdown of circ_0001742 inhibited proliferation, migration, invasion and EMT and induced apoptosis in TSCC cells. Then, miR-431-5p was identified as a target of circ_0001742, and knockdown of miR-431-5p reversed the effects of circ_0001742 knockdown on proliferation, apoptosis, migration, invasion and EMT of TSCC cells. Moreover, miR-431-5p could bind to ATF3, and overexpression of ATF3 rescued the effects mediated by miR-431-5p in TSCC cells. In addition, circ_0001742 regulated ATF3 expression through miR-431-5p. CONCLUSIONS: Our results demonstrated that circ_0001742 plays a tumor-promoting effect in TSCC cells by serving as a competing endogenous RNA (ceRNA) to regulate miR-431-5p/ATF3 axis, which might provide a potential therapeutic target for TSCC.


Subject(s)
Activating Transcription Factor 3/physiology , Carcinoma, Squamous Cell/physiopathology , Cell Movement/physiology , Cell Proliferation/physiology , MicroRNAs/physiology , RNA, Circular/physiology , Tongue Neoplasms/physiopathology , Activating Transcription Factor 3/biosynthesis , Apoptosis/physiology , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Disease Progression , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , MicroRNAs/biosynthesis , RNA, Circular/biosynthesis , Tongue Neoplasms/metabolism , Up-Regulation
12.
PLoS One ; 14(2): e0213081, 2019.
Article in English | MEDLINE | ID: mdl-30818334

ABSTRACT

c-Jun dimerization protein (JDP2) and Activating Transcription Factor 3 (ATF3) are closely related basic leucine zipper proteins. Transgenic mice with cardiac expression of either JDP2 or ATF3 showed maladaptive remodeling and cardiac dysfunction. Surprisingly, JDP2 knockout (KO) did not protect the heart following transverse aortic constriction (TAC). Instead, the JDP2 KO mice performed worse than their wild type (WT) counterparts. To test whether the maladaptive cardiac remodeling observed in the JDP2 KO mice is due to ATF3, ATF3 was removed in the context of JDP2 deficiency, referred as double KO mice (dKO). Mice were challenged by TAC, and followed by detailed physiological, pathological and molecular analyses. dKO mice displayed no apparent differences from WT mice under unstressed condition, except a moderate better performance in dKO male mice. Importantly, following TAC the dKO hearts showed low fibrosis levels, reduced inflammatory and hypertrophic gene expression and a significantly preserved cardiac function as compared with their WT counterparts in both genders. Consistent with these data, removing ATF3 resumed p38 activation in the JDP2 KO mice which correlates with the beneficial cardiac function. Collectively, mice with JDP2 and ATF3 double deficiency had reduced maladaptive cardiac remodeling and lower hypertrophy following TAC. As such, the worsening of the cardiac outcome found in the JDP2 KO mice is due to the elevated ATF3 expression. Simultaneous suppression of both ATF3 and JDP2 activity is highly beneficial for cardiac function in health and disease.


Subject(s)
Activating Transcription Factor 3/deficiency , Repressor Proteins/deficiency , Ventricular Remodeling/physiology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/physiology , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Female , Fibrosis , Heart/physiopathology , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Contraction/genetics , Myocardial Contraction/physiology , Myocardium/pathology , Repressor Proteins/genetics , Repressor Proteins/physiology , Ventricular Remodeling/genetics
13.
Adv Exp Med Biol ; 1099: 65-76, 2018.
Article in English | MEDLINE | ID: mdl-30306515

ABSTRACT

Peripheral nerve blockade (PNB) is advantageous for patients undergoing surgery to decrease the perioperative opioid consumptions and enhance recovery after surgery.Inadvertent local anesthetic (LA) administration into nerve fiber intrafascicularly easily results in unrecognized nerve injury. Using nerve block guidance either by ultrasound, electrical nerve stimulator, or using pressure devices does not prevent nerve damage, even though most of the nerve injury is transiently. The incidence of neurologic symptoms or neuropathy is in the range of 0.02-2.2%, and no significant difference of postoperative neurologic symptoms is found as compared with using ultrasound or guided nerve stimulator technique. However, intrafascicular lidocaine brought about macrophage migration into the damaged fascicle, Schwann cell proliferation, increased intensity of myelin basic protein, and shorten withdrawal time to mechanical stimuli. In dorsal root ganglion (DRG), intrafascicular LA injection increased the activated transcriptional factor 3 (ATF-3) and downregulated Nav1.8 (Nav1.8). In spinal dorsal horn (SDH), the microglia and astrocytes located in SDH were activated and proliferated after intrafascicular LA injection and returned to baseline gradually at the end of the month. This is a kind of neuropathic pain, so low injection pressure should be maintained, the correct needle bevel used, nerve stimulator or ultrasound guidance applied, and careful and deliberately slow injection employed as important parts of the injection technique to prevent intrafascicular LA administration-induced neuropathic pain.


Subject(s)
Anesthetics, Local/adverse effects , Nerve Block/adverse effects , Neuralgia/physiopathology , Peripheral Nerve Injuries/physiopathology , Activating Transcription Factor 3/physiology , Biomedical Research , Ganglia, Spinal/physiology , Humans , Injections , NAV1.8 Voltage-Gated Sodium Channel/physiology
14.
Chin Med J (Engl) ; 131(19): 2302-2309, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30246716

ABSTRACT

BACKGROUND: Myocardial ischemia injury is one of the leading causes of death and disability worldwide. Cardiac fibroblasts (CFs) have central roles in modulating cardiac function under pathophysiological conditions. Activating transcription factor 3 (ATF3) plays a self-protective role in counteracting CF dysfunction. However, the precise function of CF-specific ATF3 during myocardial infarction (MI) injury/repair remains incompletely understood. The aim of this study was to determine whether CF-specific ATF3 affected cardiac repair after MI. METHODS: Fifteen male C57BL/6 wild-type mice were performed with MI operation to observe the expression of ATF3 at 0, 0.5, 1.0, 3.0, and 7.0 days postoperation. Model for MI was constructed in ATF3TGfl/flCol1a2-Cre+ (CF-specific ATF3 overexpression group, n = 5) and ATF3TGfl/flCol1a2-Cre- male mice (without CF-specific ATF3 overexpression group, n = 5). In addition, five mice of ATF3TGfl/flCol1a2-Cre+ and ATF3TGfl/flCol1a2-Cre- were subjected to sham MI operation. Heart function was detected by ultrasound and left ventricular remodeling was observed by Masson staining (myocardial fibrosis area was detected by blue collagen deposition area) at the 28th day after MI surgery in ATF3TGfl/flCol1a2-Cre+ and ATF3TGfl/flCol1a2-Cre- mice received sham or MI operation. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect cell proliferation/cell cycle-related gene expression in cardiac tissue. BrdU staining was used to detect fibroblast proliferation. RESULTS: After establishment of an MI model, we found that ATF3 proteins were increased in the heart of mice after MI surgery and dominantly expressed in CFs. Genetic overexpression of ATF3 in CFs (ATF3TGfl/flCol1a2-Cre+ group) resulted in an improvement in the heart function as indicated by increased cardiac ejection fraction (41.0% vs. 30.5%, t = 8.610, P = 0.001) and increased fractional shortening (26.8% vs. 18.1%, t = 7.173, P = 0.002), which was accompanied by a decrease in cardiac scar area (23.1% vs. 11.0%, t = 8.610, P = 0.001). qRT-PCR analysis of CFs isolated from ATF3TGfl/flCol1a2-Cre+ and ATF3TGfl/flCol1a2-Cre- ischemic hearts revealed a distinct transcriptional profile in ATF3-overexpressing CFs, displaying pro-proliferation properties. BrdU-positive cells significantly increased in ATF3-overexpressing CFs than control CFs under angiotensin II stimuli (11.5% vs. 6.8%, t = 31.599, P = 0.001) or serum stimuli (31.6% vs. 20.1%, t = 31.599, P = 0.001). The 5(6)-carboxyfluorescein N-hydroxysuccinimidyl ester assay showed that the cell numbers of the P2 and P3 generations were higher in the ATF3-overexpressing CFs at 24 h (P2: 91.6% vs. 71.8%, t = 8.465, P = 0.015) and 48 h (P3: 81.6% vs. 51.1%, t = 9.029, P = 0.012) after serum stimulation. Notably, ATF3 overexpression-induced CF proliferation was clearly increased in the heart after MI injury. CONCLUSIONS: We identify that CF-specific ATF3 might contribute to be MI repair through upregulating the expression of cell cycle/proliferation-related genes and enhancing cell proliferation.


Subject(s)
Activating Transcription Factor 3/physiology , Fibroblasts/physiology , Myocardial Infarction , Ventricular Remodeling , Animals , Disease Models, Animal , Fibrosis , Male , Mice , Mice, Inbred C57BL , Myocardium
15.
Yakugaku Zasshi ; 137(11): 1315-1321, 2017.
Article in Japanese | MEDLINE | ID: mdl-29093367

ABSTRACT

Two types of cell death, necrosis and apoptosis, are defined in terms of cell death morphological features. We have been studying the mechanisms by which cell death processes are switched during the treatment of mouse tumor FM3A with anticancer, 5-fluoro-2'-deoxyuridine (FUdR): it induces original clone F28-7 to necrosis, but its sub-clone F28-7-A to apoptosis. We identified several such switch regulators of cell death: heat shock protein 90 (HSP90), lamin-B1, cytokeratin-19, and activating transcription factor 3 (ATF3), by using transcriptomic, proteomic analyses and siRNA screening. For example, the inhibition of HSP90 by its inhibitor geldanamycin in F28-7 caused a shift from necrosis to apoptosis. We also observed that the knockdown of lamin-B1, cytokeratin-19, or ATF3 expression in F28-7 resulted in a shift from necrosis to apoptosis. Recently, we used microRNA (miRNA, miR) microarray analyses to investigate the miRNA expression profiles in these sister cells. The miR-351 and miR-743a were expressed at higher levels in F28-7-A than in F28-7. Higher expression of miR-351 or miR-743a in F28-7, induced by transfecting the miR mimics, resulted in a switch of cell death mode: necrosis to apoptosis. Furthermore, transfection of an miR-351 inhibitor into F28-7-A resulted in morphological changes, and mode of cell death from apoptosis to necrosis. These findings suggest that the identified cell death regulators may have key roles in switching cell death mode. Possible mechanisms involving cell death regulators in the switch of necrosis or apoptosis are discussed. We propose a novel anticancer strategy targeting the switch regulators of necrosis or apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Deoxyuridine/analogs & derivatives , Necrosis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/physiology , Animals , Apoptosis/genetics , Benzoquinones/pharmacology , Benzoquinones/therapeutic use , Deoxyuridine/pharmacology , Deoxyuridine/therapeutic use , Gene Expression Profiling , HSP90 Heat-Shock Proteins/physiology , Lactams, Macrocyclic/pharmacology , Lactams, Macrocyclic/therapeutic use , Lamin Type B/genetics , Lamin Type B/physiology , Mice , MicroRNAs/antagonists & inhibitors , Molecular Targeted Therapy , Necrosis/genetics
16.
J Exp Med ; 214(8): 2349-2368, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28684431

ABSTRACT

Cancer-associated fibroblasts (CAFs) are important for tumor initiation and promotion. CSL, a transcriptional repressor and Notch mediator, suppresses CAF activation. Like CSL, ATF3, a stress-responsive transcriptional repressor, is down-modulated in skin cancer stromal cells, and Atf3 knockout mice develop aggressive chemically induced skin tumors with enhanced CAF activation. Even at low basal levels, ATF3 converges with CSL in global chromatin control, binding to few genomic sites at a large distance from target genes. Consistent with this mode of regulation, deletion of one such site 2 Mb upstream of IL6 induces expression of the gene. Observed changes are of translational significance, as bromodomain and extra-terminal (BET) inhibitors, unlinking activated chromatin from basic transcription, counteract the effects of ATF3 or CSL loss on global gene expression and suppress CAF tumor-promoting properties in an in vivo model of squamous cancer-stromal cell expansion. Thus, ATF3 converges with CSL in negative control of CAF activation with epigenetic changes amenable to cancer- and stroma-focused intervention.


Subject(s)
Activating Transcription Factor 3/physiology , Cancer-Associated Fibroblasts/physiology , Chromatin/physiology , Muscle Proteins/physiology , Animals , Carcinoma, Squamous Cell/physiopathology , Keratinocytes/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin Neoplasms/physiopathology
17.
Reprod Biol Endocrinol ; 15(1): 42, 2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28577574

ABSTRACT

BACKGROUND: A receptive endometrium is essential for maternal-embryonic molecular communication during implantation. However, the specific molecular regulatory mechanisms of the endometrial capacity remain poorly understood. Here, we examined activating transcription factor 3 (ATF3) expression in human endometria and the functional effect of ATF3 on embryo attachment in vitro. METHODS: Immunohistochemistry (IHC) was used to assess the ATF3 expression patterns in human endometria. Quantitative real-time PCR (qRT-PCR), western blotting and immunofluorescence (IF) studies were applied to explore ATF3 expression in Ishikawa cells upon estrogen (E2) and medroxyprogesterone acetate (MPA) treatment. qRT-PCR and western blotting were performed to inspect LIF (leukemia inhibitory factor) expression after enhancement or inhibition of ATF3, and a luciferase reporter assay and ChIP-PCR were used to confirm the regulatory mechanism of ATF3 to LIF. Endometrial epithelial capacity was assessed by an in vitro model of attachment of BeWo spheroids to Ishikawa cells. Western blotting was performed to compare the expression of ATF3 in endometrial samples of recurrent implantation failure (RIF) patients with that in samples from fertile women (FER) who had undergone no less than one successful embryo transplantation. RESULTS: ATF3 was located in human endometrial epithelial cells and stromal cells and was significantly induced by E2 and MPA in Ishikawa cells. Adenovirus-mediated overexpression of ATF3 in Ishikawa cells activated LIF promoter activity and enhanced its expression. Accordingly, the stimulation of BeWo spheroid adhesion promoted by ATF3 was inhibited by pretreatment with a specific antibody against LIF via the antibody-blocking assay. Moreover, ATF3 was aberrantly decreased in the endometria of RIF patients. CONCLUSIONS: Our findings suggest that ATF3 plays a significant role in regulating human endometrial receptivity and embryo attachment in vitro via up-regulation of leukemia inhibitory factor. TRIAL REGISTRATION: Construction and management of the Nanjing multi-center biobank. No. 2013-081-01 . Registered 10 Dec. 2013.


Subject(s)
Activating Transcription Factor 3/physiology , Embryo Implantation/genetics , Leukemia Inhibitory Factor/genetics , Abortion, Habitual/genetics , Abortion, Habitual/metabolism , Abortion, Habitual/pathology , Adult , Case-Control Studies , Cells, Cultured , Embryo Transfer , Endometrium/cytology , Endometrium/metabolism , Epithelial Cells/metabolism , Epithelial Cells/physiology , Female , Fertilization in Vitro , Humans , Leukemia Inhibitory Factor/metabolism , Up-Regulation/genetics , Young Adult
18.
Nat Commun ; 8: 14289, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28272390

ABSTRACT

Epithelial barrier function is maintained by coordination of cell proliferation and cell loss, whereas barrier dysfunction can lead to disease and organismal death. JNK signalling is a conserved stress signalling pathway activated by bacterial infection and tissue damage, often leading to apoptotic cell death and compensatory cell proliferation. Here we show that the stress inducible transcription factor ATF3 restricts JNK activity in the Drosophila midgut. ATF3 regulates JNK-dependent apoptosis and regeneration through the transcriptional regulation of the JNK antagonist, Raw. Enterocyte-specific ATF3 inactivation increases JNK activity and sensitivity to infection, a phenotype that can be rescued by Raw overexpression or JNK suppression. ATF3 depletion enhances intestinal regeneration triggered by infection, but does not compensate for the loss of enterocytes and ATF3-depleted flies succumb to infection due to intestinal barrier dysfunction. In sum, we provide a mechanism to explain how an ATF3-Raw module controls JNK signalling to maintain normal intestinal barrier function during acute infection.


Subject(s)
Activating Transcription Factor 3/physiology , Intestines/physiology , MAP Kinase Kinase 4/metabolism , Regeneration , Signal Transduction , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Apoptosis , Cell Division , Cell Proliferation , Cell Survival , Cytoskeletal Proteins/genetics , Disease Susceptibility , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Enterocytes/cytology , Female , Homeostasis , Intestines/cytology , Intestines/physiopathology , MAP Kinase Kinase 4/antagonists & inhibitors , Mitosis , Protein Kinase Inhibitors/pharmacology , Stress, Physiological , Transcription, Genetic
19.
Sci Rep ; 6: 30918, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27480204

ABSTRACT

Bone homeostasis is maintained by the sophisticated coupled actions of bone-resorbing osteoclasts and bone-forming osteoblasts. Here we identify activating transcription factor 3 (ATF3) as a pivotal transcription factor for the regulation of bone resorption and bone remodeling under a pathological condition through modulating the proliferation of osteoclast precursors. The osteoclast precursor-specific deletion of ATF3 in mice led to the prevention of receptor activator of nuclear factor-κB (RANK) ligand (RANKL)-induced bone resorption and bone loss, although neither bone volume nor osteoclastic parameter were markedly altered in these knockout mice under the physiological condition. RANKL-dependent osteoclastogenesis was impaired in vitro in ATF3-deleted bone marrow macrophages (BMM). Mechanistically, the deficiency of ATF3 impaired the RANKL-induced transient increase in cell proliferation of osteoclast precursors in bone marrow in vivo as well as of BMM in vitro. Moreover, ATF3 regulated cyclin D1 mRNA expression though modulating activator protein-1-dependent transcription in the osteoclast precursor, and the introduction of cyclin D1 significantly rescued the impairment of osteoclastogenesis in ATF3-deleted BMM. Therefore, these findings suggest that ATF3 could have a pivotal role in osteoclastogenesis and bone homeostasis though modulating cell proliferation under pathological conditions, thereby providing a target for bone diseases.


Subject(s)
Activating Transcription Factor 3/physiology , Bone Remodeling , Bone Resorption/prevention & control , Osteoclasts/cytology , RANK Ligand/adverse effects , Animals , Bone Marrow Cells/metabolism , Bone Resorption/etiology , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation , Cell Proliferation , Gene Expression Regulation , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/metabolism
20.
Am J Transplant ; 15(1): 76-87, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25359217

ABSTRACT

Activating transcription factor 3 (ATF3) is a stress-induced transcription factor that has been shown to repress inflammatory gene expression in multiple cell types and diseases. However, little is known about the roles and mechanisms of ATF3 in liver ischemia/reperfusion injury (IRI). In warm and cold liver IRI models, we showed that ATF3 deficiency significantly increased ischemia/reperfusion (IR)-stressed liver injury, as evidenced by increased serum alanine aminotransferase levels, histological liver damage, and hepatocellular apoptosis. These may correlate with inhibition of the intrahepatic nuclear factor erythroid-derived 2-related factor 2/heme oxygenase-1 (NRF2/HO-1) signaling pathway leading to enhancing Toll-like receptor 4/nuclear factor kappa beta (TLR4/NF-κB) activation, pro-inflammatory programs and macrophage/neutrophil trafficking, while simultaneously repressing anti-apoptotic molecules in ischemic liver. Interestingly, activation of NRF2/HO-1 signaling using an NRF2 activator, oltipraz (M2), during hepatic IRI-rescued ATF3 anti-inflammatory functions in ATF3-deficient mice. For in vitro studies, ATF3 ablation in lipopolysaccharide (LPS)-stimulated bone marrow-derived macrophages (BMMs) depressed levels of NRF2/HO-1 and PI3K/AKT, resulting in enhanced TLR4/NF-κB activation. Pretreatment of LPS-stimulated BMMs with M2 increased NRF2/HO-1 expression, promoted PI3K/AKT, which in turn suppressed TLR4/NF-κB-mediated proinflammatory mediators. Thus, our results first demonstrate ATF3-mediated NRF2/HO-1 signaling in the regulation of TLR4-driven inflammatory responses in IR-stressed livers. Our findings provide a rationale for a novel therapeutic strategy for managing IR-induced liver injury.


Subject(s)
Activating Transcription Factor 3/physiology , Heme Oxygenase-1/metabolism , Immunity, Innate/immunology , Liver/immunology , Membrane Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Reperfusion Injury/prevention & control , Toll-Like Receptor 4/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Heme Oxygenase-1/genetics , Liver/injuries , Liver/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...