Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Cells ; 10(12)2021 12 14.
Article in English | MEDLINE | ID: mdl-34944038

ABSTRACT

Psychological stress increases the risk of gastrointestinal (GI) tract diseases, which involve bidirectional communication of the GI and nerves systems. Acute stress leads to GI ulcers; however, the mechanism of the native cellular protection pathway, which safeguards tissue integrality and maintains GI homeostasis, remains to be investigated. In a mouse model of this study, restraint stress induced GI leakage, abnormal tight junction protein expression, and cell death of gut epithelial cells. The expression of activating transcription factor 3 (ATF3), a stress-responsive transcription factor, is upregulated in the GI tissues of stressed animals. ATF3-deficient mice displayed an exacerbated phenotype of GI injuries. These results suggested that, in response to stress, ATF3 is part of the native cellular protective pathway in the GI system, which could be a molecular target for managing psychological stress-induced GI tract diseases.


Subject(s)
Activating Transcription Factor 3/metabolism , Gastrointestinal Diseases/etiology , Restraint, Physical , Stress, Psychological/complications , Activating Transcription Factor 3/deficiency , Animals , Caspase 3/metabolism , Duodenum/drug effects , Duodenum/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gastrointestinal Diseases/blood , Gene Expression Regulation , Mice, Inbred C57BL , Mice, Knockout , Proton Pump Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Psychological/blood , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
2.
Cell Rep ; 36(12): 109706, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34551291

ABSTRACT

The serine synthesis pathway (SSP) involving metabolic enzymes phosphoglycerate dehydrogenase (PHGDH), phosphoserine aminotransferase 1 (PSAT1), and phosphoserine phosphatase (PSPH) drives intracellular serine biosynthesis and is indispensable for cancer cells to grow in serine-limiting environments. However, how SSP is regulated is not well understood. Here, we report that activating transcription factor 3 (ATF3) is crucial for transcriptional activation of SSP upon serine deprivation. ATF3 is rapidly induced by serine deprivation via a mechanism dependent on ATF4, which in turn binds to ATF4 and increases the stability of this master regulator of SSP. ATF3 also binds to the enhancers/promoters of PHGDH, PSAT1, and PSPH and recruits p300 to promote expression of these SSP genes. As a result, loss of ATF3 expression impairs serine biosynthesis and the growth of cancer cells in the serine-deprived medium or in mice fed with a serine/glycine-free diet. Interestingly, ATF3 expression positively correlates with PHGDH expression in a subset of TCGA cancer samples.


Subject(s)
Activating Transcription Factor 3/metabolism , Neoplasms/pathology , Serine/biosynthesis , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Activating Transcription Factor 4/metabolism , Animals , Biosynthetic Pathways/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , Neoplasms/metabolism , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Stability , Serine/deficiency , Transaminases/genetics , Transaminases/metabolism , Transplantation, Heterologous , p300-CBP Transcription Factors/metabolism
3.
Toxicology ; 460: 152890, 2021 08.
Article in English | MEDLINE | ID: mdl-34364923

ABSTRACT

Arsenic is a naturally occurring metalloid strongly associated with the incidence of lung cancer. Understanding the mechanisms of arsenic-induced carcinogenesis favors the development of effective interventions to reduce the incidence and mortality of lung cancer. In this study, we investigated the role of activating transcription factor 3 (ATF3) in arsenic-induced transformation of human bronchial epithelial cells. ATF3 was upregulated during chronic exposure to 0.25 µM arsenic, and loss of ATF3 promoted arsenic-induced transformation. Moreover, arsenic-transformed ATF3 knockout (ATF3 KO-AsT) cells exhibited more aggressive characteristics, including acceleration in proliferation, resistance to chemotherapy and increase in migratory capacity. RNA-seq revealed that pathways involved in inflammation, cell cycle, EMT and oncogenesis were affected due to ATF3 deficiency during chronic arsenic exposure. Further experiments confirmed the overproduction of IL-6, IL-8 and TNFα as well as enhanced phosphorylation of AKT and STAT3 in ATF3 KO-AsT cells. Our results demonstrate that ATF3 upregulated by chronic low-dose arsenic exposure represses cell transformation and acquisition of malignant characteristics through inhibiting the production of proinflammatory cytokines and activation of downstream proteins AKT and STAT3, providing a new strategy for the prevention of carcinogen-induced lung cancer.


Subject(s)
Activating Transcription Factor 3/deficiency , Arsenic/toxicity , Bronchi/metabolism , Inflammation Mediators/metabolism , Lung Neoplasms/metabolism , Respiratory Mucosa/metabolism , Activating Transcription Factor 3/genetics , Animals , Cell Line, Transformed , Female , Gene Knockout Techniques/methods , HEK293 Cells , Humans , Inflammation Mediators/antagonists & inhibitors , Lung Neoplasms/chemically induced , Lung Neoplasms/prevention & control , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays/methods
4.
Cell Death Dis ; 12(4): 387, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846304

ABSTRACT

Decidualization is a complex process involving cellular proliferation and differentiation of the endometrial stroma and is required to establish and support pregnancy. Dysregulated decidualization has been reported to be a critical cause of recurrent implantation failure (RIF). In this study, we found that Activating transcription factor 3 (ATF3) expression was significantly downregulated in the endometrium of RIF patients. Knockdown of ATF3 in human endometrium stromal cells (hESCs) hampers decidualization, while overexpression could trigger the expression of decidual marker genes, and ameliorate the decidualization of hESCs from RIF patients. Mechanistically, ATF3 promotes decidualization by upregulating FOXO1 via suppressing miR-135b expression. In addition, the endometrium of RIF patients was hyperproliferative, while overexpression of ATF3 inhibited the proliferation of hESCs through CDKN1A. These data demonstrate the critical roles of endometrial ATF3 in regulating decidualization and proliferation, and dysregulation of ATF3 in the endometrium may be a novel cause of RIF and therefore represent a potential therapeutic target for RIF.


Subject(s)
Activating Transcription Factor 3/deficiency , Embryo Implantation/physiology , Endometrium/physiology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Cell Proliferation/physiology , Cells, Cultured , Decidua/metabolism , Endometrium/metabolism , Female , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Stromal Cells/metabolism , Transfection
5.
Commun Biol ; 2: 389, 2019.
Article in English | MEDLINE | ID: mdl-31667363

ABSTRACT

Billions of people have obesity-related metabolic syndromes such as diabetes and hyperlipidemia. Promoting the browning of white adipose tissue has been suggested as a potential strategy, but a drug still needs to be identified. Here, genetic deletion of activating transcription factor 3 (ATF3-/- ) in mice under a high-fat diet (HFD) resulted in obesity and insulin resistance, which was abrogated by virus-mediated ATF3 restoration. ST32da, a synthetic ATF3 inducer isolated from Salvia miltiorrhiza, promoted ATF3 expression to downregulate adipokine genes and induce adipocyte browning by suppressing the carbohydrate-responsive element-binding protein-stearoyl-CoA desaturase-1 axis. Furthermore, ST32da increased white adipose tissue browning and reduced lipogenesis in HFD-induced obese mice. The anti-obesity efficacy of oral ST32da administration was similar to that of the clinical drug orlistat. Our study identified the ATF3 inducer ST32da as a promising therapeutic drug for treating diet-induced obesity and related metabolic disorders.


Subject(s)
Activating Transcription Factor 3/metabolism , Adipocytes, Brown/metabolism , Obesity/metabolism , 3T3-L1 Cells , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Adipocytes, Brown/pathology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Anti-Obesity Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Body Temperature Regulation/physiology , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Insulin Resistance , Lipogenesis/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/prevention & control , Orlistat/pharmacology , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Salvia miltiorrhiza/chemistry
6.
eNeuro ; 6(2)2019.
Article in English | MEDLINE | ID: mdl-30993183

ABSTRACT

Genome editing techniques have facilitated significant advances in our understanding of fundamental biological processes, and the Cre-Lox system has been instrumental in these achievements. Driving Cre expression specifically in injured neurons has not been previously possible: we sought to address this limitation in mice using a Cre-ERT2 construct driven by a reliable indicator of axotomy, activating transcription factor 3 (ATF3). When crossed with reporter mice, a significant amount of recombination was achieved (without tamoxifen treatment) in peripherally-projecting sensory, sympathetic, and motoneurons after peripheral nerve crush in hemizygotes (65-80% by 16 d) and was absent in uninjured neurons. Importantly, injury-induced recombination did not occur in Schwann cells distal to the injury, and with a knock-out-validated antibody we verified an absence of ATF3 expression. Functional recovery following sciatic nerve crush in ATF3-deficient mice (both hemizygotes and homozygotes) was delayed, indicating previously unreported haploinsufficiency. In a proof-of-principle experiment, we crossed the ATF3-CreERT2 line with a floxed phosphatase and tensin homolog (PTEN) line and show significantly improved axonal regeneration, as well as more complete recovery of neuromuscular function. We also demonstrate the utility of the ATF3-CreERT2 hemizygous line by characterizing recombination after lateral spinal hemisection (C8/T1), which identified specific populations of ascending spinal cord neurons (including putative spinothalamic and spinocerebellar) and descending supraspinal neurons (rubrospinal, vestibulospinal, reticulospinal and hypothalamic). We anticipate these mice will be valuable in distinguishing axotomized from uninjured neurons of several different classes (e.g., via reporter expression), and in probing the function of any number of genes as they relate to neuronal injury and regeneration.


Subject(s)
Activating Transcription Factor 3/metabolism , Gene Editing/methods , Gene Expression Regulation , Nerve Regeneration , Peripheral Nerve Injuries , Recovery of Function , Schwann Cells , Sensory Receptor Cells , Spinal Cord Injuries , Activating Transcription Factor 3/deficiency , Animals , Axotomy , Behavior, Animal/physiology , Disease Models, Animal , Feasibility Studies , Gene Expression/genetics , Integrases/metabolism , Mice , Mice, Transgenic , PTEN Phosphohydrolase/metabolism , Proof of Concept Study , Schwann Cells/metabolism , Sciatic Nerve/injuries , Sensory Receptor Cells/metabolism
7.
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
8.
Cell Stem Cell ; 23(2): 210-225.e6, 2018 Aug 02.
Article in English | MEDLINE | ID: mdl-30075129

ABSTRACT

The cellular and mechanistic bases underlying endothelial regeneration of adult large vessels have proven challenging to study. Using a reproducible in vivo aortic endothelial injury model, we characterized cellular dynamics underlying the regenerative process through a combination of multi-color lineage tracing, parabiosis, and single-cell transcriptomics. We found that regeneration is a biphasic process driven by distinct populations arising from differentiated endothelial cells. The majority of cells immediately adjacent to the injury site re-enter the cell cycle during the initial damage response, with a second phase driven by a highly proliferative subpopulation. Endothelial regeneration requires activation of stress response genes including Atf3, and aged aortas compromised in their reparative capacity express less Atf3. Deletion of Atf3 reduced endothelial proliferation and compromised the regeneration. These findings provide important insights into cellular dynamics and mechanisms that drive responses to large vessel injury.


Subject(s)
Aorta/cytology , Endothelial Cells/cytology , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/metabolism , Animals , Aorta/injuries , Aorta/metabolism , Cell Proliferation , Endothelial Cells/metabolism , Kinetics , Mice , Mice, Inbred C57BL
9.
Toxicol Lett ; 287: 113-121, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29378244

ABSTRACT

Airborne particulate matter (PM) has been reported to be associated with a wide range of respiratory disorders. However, the mechanisms underlying PM-induced airway inflammation remain largely unknown. Generally, ATF3 negatively regulates pro-inflammatory cytokines production in response to TLR4 signaling. Here we first showed ATF3 has promoting effects in PM-induced airway inflammation in vitro an in vivo. We demonstrated PM significantly upregulated ATF3 expression in HBE cells and in mouse lung tissues. ATF3 siRNA markedly inhibited, while ATF3-recombinant over-expression plasmid significantly increased PM-induced IL-6 expression in cultured HBE cells, and PM-induced IL-6, CXCL2 expression as well as neutrophil infiltration, mucus over-production in the lung of ATF3-/- mice were all notably reduced relative to the wild-type littermates. Furthermore, we showed ATF3 mediated PM-induced inflammatory cytokines expression partly through NF-κB and AP-1 pathways. Our data further elucidates the mechanisms underlying PM-induced airway inflammation, and indicates ATF3 may function as different role in response to different stimuli.


Subject(s)
Activating Transcription Factor 3/metabolism , Bronchi/drug effects , Epithelial Cells/drug effects , Particulate Matter/toxicity , Pneumonia/metabolism , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Animals , Bronchi/metabolism , Bronchi/pathology , Cell Line , Chemokine CXCL2/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Interleukin-6/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mucus/metabolism , NF-kappa B/metabolism , Neutrophil Infiltration/drug effects , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/pathology , RNA Interference , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Transfection
10.
Circulation ; 135(21): 2041-2057, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28249877

ABSTRACT

BACKGROUND: Hypertensive ventricular remodeling is a common cause of heart failure. However, the molecular mechanisms regulating ventricular remodeling remain poorly understood. METHODS: We used a discovery-driven/nonbiased approach to identify increased activating transcription factor 3 (ATF3) expression in hypertensive heart. We used loss/gain of function approaches to understand the role of ATF3 in heart failure. We also examined the mechanisms through transcriptome, chromatin immunoprecipitation sequencing analysis, and in vivo and in vitro experiments. RESULTS: ATF3 expression increased in murine hypertensive heart and human hypertrophic heart. Cardiac fibroblast cells are the primary cell type expressing high ATF3 levels in response to hypertensive stimuli. ATF3 knockout (ATF3KO) markedly exaggerated hypertensive ventricular remodeling, a state rescued by lentivirus-mediated/miRNA-aided cardiac fibroblast-selective ATF3 overexpression. Conversely, conditional cardiac fibroblast cell-specific ATF3 transgenic overexpression significantly ameliorated ventricular remodeling and heart failure. We identified Map2K3 as a novel ATF3 target. ATF3 binds with the Map2K3 promoter, recruiting HDAC1, resulting in Map2K3 gene-associated histone deacetylation, thereby inhibiting Map2K3 expression. Genetic Map2K3 knockdown rescued the profibrotic/hypertrophic phenotype in ATF3KO cells. Last, we demonstrated that p38 is the downstream molecule of Map2K3 mediating the profibrotic/hypertrophic effects in ATF3KO animals. Inhibition of p38 signaling reduced transforming growth factor-ß signaling-related profibrotic and hypertrophic gene expression, and blocked exaggerated cardiac remodeling in ATF3KO cells. CONCLUSIONS: Our study provides the first evidence that ATF3 upregulation in cardiac fibroblasts in response to hypertensive stimuli protects the heart by suppressing Map2K3 expression and subsequent p38-transforming growth factor-ß signaling. These results suggest that positive modulation of cardiac fibroblast ATF3 may represent a novel therapeutic approach against hypertensive cardiac remodeling.


Subject(s)
Activating Transcription Factor 3/metabolism , Fibroblasts/enzymology , Heart Failure/prevention & control , Hypertrophy, Left Ventricular/prevention & control , MAP Kinase Kinase 3/metabolism , Myocardium/enzymology , Ventricular Function, Left , Ventricular Remodeling , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylation , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Angiotensin II , Animals , Binding Sites , Cells, Cultured , Disease Models, Animal , Fibroblasts/pathology , Fibrosis , Genetic Predisposition to Disease , Heart Failure/enzymology , Heart Failure/etiology , Heart Failure/physiopathology , Histone Deacetylase 1/metabolism , Histones/metabolism , Humans , Hypertension/chemically induced , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/physiopathology , MAP Kinase Kinase 3/genetics , Male , Mice, Knockout , Myocardium/pathology , Phenotype , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Time Factors , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
11.
J Cell Physiol ; 232(12): 3727-3734, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28177121

ABSTRACT

Excessive inflammatory responses are critical in the pathogenesis of acute lung injury (ALI). Activating transcription factor 3 (ATF3) is a stress-induced transcriptional regulator that is a negative regulator of inflammatory responses. Therefore, we investigated the role and signaling pathways of ATF3 in lipopolysaccharide (LPS)-induced ALI in mice. The mouse macrophage RAW264.7 cells were cultured on HTS 24-Transwell filter plates in presence of ATF3 siRNA before exposure to LPS. ATF3 knock-out (KO) and wild type (WT) mice were challenged by intra-peritoneal injection of LPS (15 mg/kg). Gene analysis was used to analyze differential gene expression between ATF3 KO and WT mice. LPS increased the expression of ATF3 in RAW264.7 cells and in lung tissues of mice, The concentration of TNFα and IL-6 was significantly increased in ATF3 siRNA-treated RAW264.7 cells compared to control cells after LPS stimulation. The concentration of TNFα, IL-6 and IL-1ß in serum and lung tissue of ATF3 KO mice was significantly increased compared to ATF3 WT mice. In addition, the lung wet/dry weight and BALF protein were significantly increased in ATF3 KO mice after LPS injection at 6, 24, and 48 hr. The survival of ATF3 KO mice significantly decreased. Differential gene analysis showed that TL1A was highly expressed in LPS-induced lung tissues of ATF3 KO mice.Moreover, ATF3 down-regulated the expression of TL1A in RAW264.7 cells and in lung tissues. These findings suggest that ATF3 protects against LPS-induced ALI via inhibiting TL1A expression.


Subject(s)
Activating Transcription Factor 3/metabolism , Acute Lung Injury/prevention & control , Lipopolysaccharides , Lung/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , Capillary Permeability , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Down-Regulation , Gene Expression Profiling/methods , Genetic Predisposition to Disease , Inflammation Mediators/metabolism , Lung/blood supply , Lung/pathology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , RAW 264.7 Cells , RNA Interference , Signal Transduction , Time Factors , Transfection , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
12.
Cardiovasc Res ; 113(2): 134-146, 2017 02.
Article in English | MEDLINE | ID: mdl-28082453

ABSTRACT

AIMS: Obesity and type 2 diabetes (T2D) trigger a harmful stress-induced cardiac remodeling process known as cardiomyopathy. These diseases represent a serious and widespread health problem in the Western world; however the underlying molecular basis is not clear. ATF3 is an 'immediate early' gene whose expression is highly and transiently induced in response to multiple stressors such as metabolic, oxidative, endoplasmic reticulum and inflammation, stressors that are involved in T2D cardiomyopathy. The role of ATF3 in diabetic cardiomyopathy is currently unknown. Our research has aimed to study the effect of ATF3 expression on cardiomyocytes, heart function and glucose homeostasis in an obesity-induced T2D mouse model. METHODS AND RESULTS: We used wild type mice (WT) as well as mutant mice with a cardiac-specific ATF3 deficiency (ATF3-cKO). Mice were fed a high-fat diet (HFD) for 15 weeks. HFD induced high ATF3 expression in cardiomyocytes. Mice were examined for cardiac remodeling processes and the diabetic state was assessed. HFD-fed ATF3-cKO mice exhibited severe cardiac fibrosis, higher levels of heart hypertrophic markers, increased inflammation and worse cardiac function, as compared to WT mice. Interestingly, HFD-fed ATF3-cKO mice display increased hyperglycemia and reduced glucose tolerance, despite higher blood insulin levels, as compared to HFD-fed WT mice. Elevated levels of the cardiac inflammatory cytokines IL-6 and TNFα leading to impaired insulin signalling may partially explain the peripheral glucose intolerance. CONCLUSIONS: Cardiac ATF3 has a protective role in dampening the HFD-induced cardiac remodeling processes. ATF3 exerts both local and systemic effects related to T2D-induced cardiomyopathy. This study provides a strong relationship between heart remodeling processes and blood glucose homeostasis.


Subject(s)
Activating Transcription Factor 3/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetic Cardiomyopathies/blood , Myocytes, Cardiac/metabolism , Ventricular Remodeling , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Animals , Cardiomegaly/blood , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Diabetes Mellitus, Type 2/etiology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Diet, High-Fat , Disease Models, Animal , Fatty Acids, Nonesterified/pharmacology , Fibrosis , Genetic Predisposition to Disease , Homeostasis , Inflammation Mediators/metabolism , Insulin/blood , Integrases/genetics , Interleukin-6/blood , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Phenotype , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/blood , Ventricular Remodeling/drug effects
16.
Am J Physiol Heart Circ Physiol ; 309(8): H1326-35, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26342068

ABSTRACT

Numerous fibrotic and inflammatory changes occur in the failing heart. Recent evidence indicates that certain transcription factors, such as activating transcription factor 3 (ATF3), are activated during heart failure. Because ATF3 may be upregulated in the failing heart and affect inflammation, we focused on the potential role of ATF3 on postinfarct heart failure. We subjected anesthetized, wild-type mice to nonreperfused myocardial infarction and observed a significant induction in ATF3 expression and nuclear translocation. To test whether the induction of ATF3 affected the severity of heart failure, we subjected wild-type and ATF3-null mice to nonreperfused infarct-induced heart failure. There were no differences in cardiac function between the two genotypes, except at the 2-wk time point; however, ATF3-null mice survived the heart failure protocol at a significantly higher rate than the wild-type mice. Similar to the slight favorable improvements in chamber dimensions at 2 wk, we also observed greater cardiomyocyte hypertrophy and more fibrosis in the noninfarcted regions of the ATF3-null hearts compared with the wild-type. Nevertheless, there were no significant group differences at 4 wk. Furthermore, we found no significant differences in markers of inflammation between the wild-type and ATF3-null hearts. Our data suggest that ATF3 suppresses fibrosis early but not late during infarct-induced heart failure. Although ATF3 deficiency was associated with more fibrosis, this did not occur at the expense of survival, which was higher in the ATF3-null mice. Overall, ATF3 may serve a largely maladaptive role during heart failure.


Subject(s)
Activating Transcription Factor 3/metabolism , Heart Failure/etiology , Myocardial Infarction/complications , Myocardium/metabolism , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Animals , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Disease Models, Animal , Fibrosis , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Time Factors , Ventricular Remodeling
18.
Int J Cardiol ; 198: 232-40, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26201690

ABSTRACT

RATIONALE: Pressure overload induces adaptive remodeling processes in the heart. However, when pressure overload persists, adaptive changes turn into maladaptive alterations leading to cardiac hypertrophy and heart failure. ATF3 is a stress inducible transcription factor that is transiently expressed following neuroendocrine stimulation. However, its role in chronic pressure overload dependent cardiac hypertrophy is currently unknown. OBJECTIVE: The objective of the study was to study the role of ATF3 in chronic pressure overload dependent cardiac remodeling processes. METHODS AND RESULTS: Pressure overload was induced by phenylephrine (PE) mini-osmotic pumps in various mice models of whole body, cardiac specific, bone marrow (BM) specific and macrophage specific ATF3 ablations. We show that ATF3-KO mice exhibit a significantly reduced expression of cardiac remodeling markers following chronic pressure overload. Consistently, the lack of ATF3 specifically in either cardiomyocytes or BM derived cells blunts the hypertrophic response to PE infusion. A unique cross-talk between cardiomyocytes and macrophages was identified. Cardiomyocytes induce an ATF3 dependent induction of an inflammatory response leading to macrophage recruitment to the heart. Adoptive transfer of wild type macrophages, but not ATF3-KO derived macrophages, into wild type mice potentiates maladaptive response to PE infusion. CONCLUSIONS: Collectively, this study places ATF3 as a key regulator in promoting pressure overload induced cardiac hypertrophy through a cross-talk between cardiomyocytes and macrophages. Inhibiting this cross-talk may serve as a useful approach to blunt maladaptive remodeling processes in the heart.


Subject(s)
Activating Transcription Factor 3/deficiency , Cardiomegaly/metabolism , Macrophages/metabolism , Myocytes, Cardiac/metabolism , Ventricular Remodeling/physiology , Activating Transcription Factor 3/biosynthesis , Animals , Cardiomegaly/pathology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology
19.
J Cell Physiol ; 229(9): 1202-11, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24420912

ABSTRACT

Transcriptional repressor activating transcription factor 3 (ATF3) is induced by various stress stimuli, including inflammation-induced renal injury. In addition, ATF3 also down-regulates adhesion molecules like intercellular adhesion molecule (ICAM), vascular cell adhesion molecule (VCAM), and monocyte chemotactic protein-1 (MCP-1). However, the relation between up-regulated ATF3 after renal ischemia/reperfusion (I/R) injury and MCP-1 is not completely understood. In this study, we demonstrated that, in renal I/R induced inflammation, induction of adhesion molecules (interleukin-6, P-selectin, E-selectin, ICAM, VCAM, and MCP-1) was higher in ATF3-knockout mice than in wild-type animals. Molecular and biochemical analyses revealed that ATF3 binds to the ATF/CRE sites in the MCP-1 promoter and inhibits the secretion of MCP-1 from renal epithelial cells after I/R injury. Urinary exosome containing ATF3 RNA was 60-fold higher in patients with acute kidney injury than in normal controls, but no difference in total urinary ATF3 RNA levels was found. In addition, in vitro study showed that exosome containing ATF3 RNA derived from epithelial cells also inhibits MCP-1 expression in the epithelial cells and macrophage migration. Furthermore, direct administration of the epithelium-derived exosomal ATF3 RNA attenuates I/R induced kidney injury. Together, our studies reveal a novel regulatory mechanism of MCP-1 expression mediated by the exosomal ATF3 RNA under renal I/R insult and suggest a potential targeted therapy for I/R induced acute kidney injury.


Subject(s)
Activating Transcription Factor 3/metabolism , Acute Kidney Injury/metabolism , Chemokine CCL2/metabolism , Exosomes/metabolism , Inflammation Mediators/metabolism , Kidney/metabolism , Reperfusion Injury/metabolism , Transcription, Genetic , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/urine , Acute Kidney Injury/genetics , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Acute Kidney Injury/urine , Adult , Aged , Aged, 80 and over , Animals , Binding Sites , Cell Line , Chemokine CCL2/genetics , Disease Models, Animal , Down-Regulation , Epithelial Cells/immunology , Epithelial Cells/metabolism , Exosomes/immunology , Female , Humans , Kidney/blood supply , Kidney/immunology , Kidney/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Rats , Reperfusion Injury/genetics , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Reperfusion Injury/urine , Signal Transduction , Time Factors , Transfection
20.
Arterioscler Thromb Vasc Biol ; 33(9): 2088-96, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23868936

ABSTRACT

OBJECTIVE: Elevation of triglyceride-rich lipoproteins (TGRLs) contributes to the risk of atherosclerotic cardiovascular disease. Our work has shown that TGRL lipolysis products in high physiological to pathophysiological concentrations cause endothelial cell injury; however, the mechanisms remain to be delineated. APPROACH AND RESULTS: We analyzed the transcriptional signaling networks in arterial endothelial cells exposed to TGRL lipolysis products. When human aortic endothelial cells in culture were exposed to TGRL lipolysis products, activating transcription factor 3 (ATF3) was identified as a principal response gene. Induction of ATF3 mRNA and protein was confirmed by quantitative reverse-transcription polymerase chain reaction and Western blot respectively. Immunofluorescence analysis showed that ATF3 accumulated in the nuclei of cells treated with lipolysis products. Nuclear expression of phosphorylated c-Jun N-terminal kinase (JNK), previously shown to be an initiator of the ATF3 signaling cascade, also was demonstrated. Small interfering RNA (siRNA)-mediated inhibition of ATF3 blocked lipolysis products-induced transcription of E-selectin and interleukin-8, but not interleukin-6 or nuclear factor-κB. c-Jun, a downstream protein in the JNK pathway, was phosphorylated, whereas expression of nuclear factor-κB-dependent JunB was downregulated. Additionally, JNK siRNA suppressed ATF3 and p-c-Jun protein expression, suggesting that JNK is upstream of the ATF3 signaling pathway. In vivo studies demonstrated that infusion of TGRL lipolysis products into wild-type mice induced nuclear ATF3 accumulation in carotid artery endothelium. ATF3(-/-) mice were resistant to vascular apoptosis precipitated by treatment with TGRL lipolysis products. Also peripheral blood monocytes isolated from postprandial humans had increased ATF3 expression as compared with fasting monocytes. CONCLUSIONS: This study demonstrates that TGRL lipolysis products activate ATF3-JNK transcription factor networks and induce endothelial cells inflammatory response.


Subject(s)
Activating Transcription Factor 3/metabolism , Apoptosis , Endothelial Cells/metabolism , Inflammation/metabolism , Lipoproteins/metabolism , Triglycerides/metabolism , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Animals , Blotting, Western , Cells, Cultured , E-Selectin/metabolism , Endothelial Cells/immunology , Endothelial Cells/pathology , Enzyme Activation , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/metabolism , Interleukin-8/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Leukocytes, Mononuclear/metabolism , Lipolysis , Lipoprotein Lipase/metabolism , Lipoproteins/blood , Lipoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phosphorylation , RNA Interference , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL