Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
1.
Tissue Cell ; 89: 102447, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38991270

ABSTRACT

Activating transcription factor 3 (ATF3) has been identified as a regulator associated with osteoblast differentiation. However, the effects of ATF3 on the osteogenic differentiation and proliferation of human periodontal stem cells (hPDLSCs) in periodontitis have not been reported. With the purpose of establishing an in vitro model of periodontitis, hPDLSCs were challenged with lipopolysaccharide (LPS). The Cell Counting Kit-8 assay was applied to assess cell viability, while reverse transcription-quantitative PCR and western blotting were employed to detect ATF3 expression. Inflammatory release was assessed using ELISA, together with western blotting. Lipid peroxidation was explored using the C11 BODIPY 581/591 probe, biochemical kits, thiobarbituric acid reactive substances (TBARS) assay and DCFH-DA staining. Iron and Fe2+ levels, and the expression levels of ferroptosis-related proteins were measured using corresponding kits and western blotting. Osteogenic differentiative capability was evaluated using alkaline phosphatase staining, Alizarin red staining and western blotting. The expression levels of proteins associated with Nrf2/HO-1 signaling were identified using western blotting. The results indicated that ATF3 expression was upregulated in LPS-induced hPDLSCs. The knockdown of ATF3 alleviated the LPS-induced inflammatory response in hPDLSCs, together with increased levels of TNF-α, IL-6, IL-1ß, Cox-2 and iNOS, and decreased levels of IL-10. ATF3 silencing also led to lower TBARS production rate, and reduced levels of reactive oxygen species, iron, Fe2+, ACSL4 and TFR1, whereas it elevated the levels of SLC7A11 and GPX4. In addition, ATF3 silencing promoted hPDLSC mineralization and cell differentiation, and elevated the levels of OCN2, RUNX2 and BMP2. Additionally, ATF3 depletion upregulated the expression levels of proteins related with Nrf2/HO-1 signaling. The Nrf2 inhibitor ML385 partially counteracted the effects of ATF3 interference on the LPS-challenged inflammatory response, lipid peroxidation, ferroptosis as well as osteogenic differentiative capability in hPDLSCs. In summary, the results revealed that ATF3 silencing suppressed inflammation and ferroptosis, while it improved osteogenic differentiation in LPS-induced hPDLSCs by regulating Nrf2/HO-1 signaling, which may provide promising therapeutic targets for the treatment of periodontitis.

2.
Int J Mol Sci ; 25(10)2024 May 11.
Article in English | MEDLINE | ID: mdl-38791301

ABSTRACT

Psychological stress increases risk of gastrointestinal tract diseases. However, the mechanism behind stress-induced gastrointestinal injury is not well understood. The objective of our study is to elucidate the putative mechanism of stress-induced gastrointestinal injury and develop an intervention strategy. To achieve this, we employed the restraint stress mouse model, a well-established method to study the pathophysiological changes associated with psychological stress in mice. By orally administering gut-nonabsorbable Evans blue dye and monitoring its plasma levels, we were able to track the progression of gastrointestinal injury in live mice. Additionally, flow cytometry was utilized to assess the viability, death, and inflammatory status of splenic leukocytes, providing insights into the stress-induced impact on the innate immune system associated with stress-induced gastrointestinal injury. Our findings reveal that neutrophils represent the primary innate immune leukocyte lineage responsible for stress-induced inflammation. Splenic neutrophils exhibited elevated expression levels of the pro-inflammatory cytokine IL-1, cellular reactive oxygen species, mitochondrial burden, and cell death following stress challenge compared to other innate immune cells such as macrophages, monocytes, and dendritic cells. Regulated cell death analysis indicated that NETosis is the predominant stress-induced cell death response among other analyzed regulated cell death pathways. NETosis culminates in the formation and release of neutrophil extracellular traps, which play a crucial role in modulating inflammation by binding to pathogens. Treatment with the NETosis inhibitor GSK484 rescued stress-induced neutrophil extracellular trap release and gastrointestinal injury, highlighting the involvement of neutrophil extracellular traps in stress-induced gastrointestinal inflammation. Our results suggest that neutrophil NETosis could serve as a promising drug target for managing psychological stress-induced gastrointestinal injuries.


Subject(s)
Inflammation , Neutrophils , Restraint, Physical , Stress, Psychological , Animals , Mice , Neutrophils/immunology , Neutrophils/metabolism , Stress, Psychological/complications , Stress, Psychological/immunology , Inflammation/pathology , Male , Mice, Inbred C57BL , Extracellular Traps/metabolism , Gastrointestinal Diseases/etiology , Disease Models, Animal , Reactive Oxygen Species/metabolism
3.
Clin Transl Med ; 14(4): e1650, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38649772

ABSTRACT

BACKGROUND: Although many molecules have been investigated as biomarkers for spinal cord injury (SCI) or ischemic stroke, none of them are specifically induced in central nervous system (CNS) neurons following injuries with low baseline expression. However, neuronal injury constitutes a major pathology associated with SCI or stroke and strongly correlates with neurological outcomes. Biomarkers characterized by low baseline expression and specific induction in neurons post-injury are likely to better correlate with injury severity and recovery, demonstrating higher sensitivity and specificity for CNS injuries compared to non-neuronal markers or pan-neuronal markers with constitutive expressions. METHODS: In animal studies, young adult wildtype and global Atf3 knockout mice underwent unilateral cervical 5 (C5) SCI or permanent distal middle cerebral artery occlusion (pMCAO). Gene expression was assessed using RNA-sequencing and qRT-PCR, while protein expression was detected through immunostaining. Serum ATF3 levels in animal models and clinical human samples were measured using commercially available enzyme-linked immune-sorbent assay (ELISA) kits. RESULTS: Activating transcription factor 3 (ATF3), a molecular marker for injured dorsal root ganglion sensory neurons in the peripheral nervous system, was not expressed in spinal cord or cortex of naïve mice but was induced specifically in neurons of the spinal cord or cortex within 1 day after SCI or ischemic stroke, respectively. Additionally, ATF3 protein levels in mouse blood significantly increased 1 day after SCI or ischemic stroke. Importantly, ATF3 protein levels in human serum were elevated in clinical patients within 24 hours after SCI or ischemic stroke. Moreover, Atf3 knockout mice, compared to the wildtype mice, exhibited worse neurological outcomes and larger damage regions after SCI or ischemic stroke, indicating that ATF3 has a neuroprotective function. CONCLUSIONS: ATF3 is an easily measurable, neuron-specific biomarker for clinical SCI and ischemic stroke, with neuroprotective properties. HIGHLIGHTS: ATF3 was induced specifically in neurons of the spinal cord or cortex within 1 day after SCI or ischemic stroke, respectively. Serum ATF3 protein levels are elevated in clinical patients within 24 hours after SCI or ischemic stroke. ATF3 exhibits neuroprotective properties, as evidenced by the worse neurological outcomes and larger damage regions observed in Atf3 knockout mice compared to wildtype mice following SCI or ischemic stroke.


Subject(s)
Activating Transcription Factor 3 , Biomarkers , Ischemic Stroke , Neurons , Spinal Cord Injuries , Animals , Female , Humans , Male , Mice , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 3/genetics , Biomarkers/metabolism , Biomarkers/blood , Disease Models, Animal , Ischemic Stroke/metabolism , Ischemic Stroke/genetics , Ischemic Stroke/blood , Mice, Knockout , Neurons/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/genetics , Spinal Cord Injuries/complications
4.
Cell Commun Signal ; 22(1): 240, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664711

ABSTRACT

BACKGROUND: The repair of peripheral nerve injury poses a clinical challenge, necessitating further investigation into novel therapeutic approaches. In recent years, bone marrow mesenchymal stromal cell (MSC)-derived mitochondrial transfer has emerged as a promising therapy for cellular injury, with reported applications in central nerve injury. However, its potential therapeutic effect on peripheral nerve injury remains unclear. METHODS: We established a mouse sciatic nerve crush injury model. Mitochondria extracted from MSCs were intraneurally injected into the injured sciatic nerves. Axonal regeneration was observed through whole-mount nerve imaging. The dorsal root ganglions (DRGs) corresponding to the injured nerve were harvested to test the gene expression, reactive oxygen species (ROS) levels, as well as the degree and location of DNA double strand breaks (DSBs). RESULTS: The in vivo experiments showed that the mitochondrial injection therapy effectively promoted axon regeneration in injured sciatic nerves. Four days after injection of fluorescently labeled mitochondria into the injured nerves, fluorescently labeled mitochondria were detected in the corresponding DRGs. RNA-seq and qPCR results showed that the mitochondrial injection therapy enhanced the expression of Atf3 and other regeneration-associated genes in DRG neurons. Knocking down of Atf3 in DRGs by siRNA could diminish the therapeutic effect of mitochondrial injection. Subsequent experiments showed that mitochondrial injection therapy could increase the levels of ROS and DSBs in injury-associated DRG neurons, with this increase being correlated with Atf3 expression. ChIP and Co-IP experiments revealed an elevation of DSB levels within the transcription initiation region of the Atf3 gene following mitochondrial injection therapy, while also demonstrating a spatial proximity between mitochondria-induced DSBs and CTCF binding sites. CONCLUSION: These findings suggest that MSC-derived mitochondria injected into the injured nerves can be retrogradely transferred to DRG neuron somas via axoplasmic transport, and increase the DSBs at the transcription initiation regions of the Atf3 gene through ROS accumulation, which rapidly release the CTCF-mediated topological constraints on chromatin interactions. This process may enhance spatial interactions between the Atf3 promoter and enhancer, ultimately promoting Atf3 expression. The up-regulation of Atf3 induced by mitochondria further promotes the expression of downstream regeneration-associated genes and facilitates axon regeneration.


Subject(s)
Activating Transcription Factor 3 , Axons , DNA Breaks, Double-Stranded , Ganglia, Spinal , Mesenchymal Stem Cells , Mitochondria , Nerve Regeneration , Reactive Oxygen Species , Sciatic Nerve , Up-Regulation , Animals , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Reactive Oxygen Species/metabolism , Axons/metabolism , Nerve Regeneration/genetics , Up-Regulation/genetics , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Ganglia, Spinal/metabolism , Mice, Inbred C57BL , Male
5.
Int J Immunopathol Pharmacol ; 38: 3946320241227320, 2024.
Article in English | MEDLINE | ID: mdl-38248871

ABSTRACT

Background: Clear cell renal cell carcinoma (ccRCC) is the most invasive type of cancer, with a high risk of metastasis and recurrence. Therefore, there is an urgent need to identify novel prognostic predictors and therapeutic targets of ccRCC. Activating transcription factor 3 (ATF3), a tumor oncogene or repressor, has rarely been examined in ccRCC. In the present study, we comprehensively elucidate the prognostic value and potential functions of ATF3 in ccRCC.Methods: Several TCGA-based online databases were used to analyze ATF3 expression in ccRCC and determine ccRCC prognosis. The upstream-binding micro (mi) RNAs of ATF3 and long non-coding (lnc)RNAs were predicted using the StarBase database.Results: Analysis of several TCGA-based online databases showed that ATF3 expression is decreased in ccRCC, suggesting a significant association with the prognosis of patients with ccRCC. Furthermore, we found hsa-miR-221-3p to be potential regulatory miRNA of ATF3 in ccRCC. Prediction and analysis of the upstream lncRNAs indicated that PAXIP1-AS2 and OIP5-AS1 were the most potent upstream lncRNAs of the hsa-miR-221-3p/ATF3 axis in ccRCC. The results of the GO and KEGG analyses implied that ATF3 is likely involved in the regulation of apoptotic signaling in response to endoplasmic reticulum (ER) stress in ccRCC. Correlation analysis revealed a positive relationship between ATF3 expression and ER stress.Conclusions: Our in silico findings highlighted that ATF3 expression was low in ccRCC and negatively correlated with poor prognosis. Furthermore, PAXIP1-AS2 and the OIP5-AS1/hsa-miR-221-3p/ATF3 axis were identified as significant potential regulators of ER stress-mediated apoptosis in ccRCC.


Subject(s)
Activating Transcription Factor 3 , Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Activating Transcription Factor 3/genetics , Biomarkers , Carcinoma , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/diagnosis , Kidney Neoplasms/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics
6.
Autophagy ; 20(1): 29-44, 2024 01.
Article in English | MEDLINE | ID: mdl-37561040

ABSTRACT

ABBREVIATIONS: AAV: adeno-associated virus; ATF3: activating transcription factor 3; ATG7: autophagy related 7; AVIL: advillin; cADPR: cyclic ADP ribose; CALC: calcitonin/calcitonin-related polypeptide; CMT: Charcot-Marie-Tooth disease; cKO: conditional knockout; DEG: differentially expressed gene; DRG: dorsal root ganglion; FE-SEM: field emission scanning electron microscopy; IF: immunofluorescence; NCV: nerve conduction velocity; PVALB: parvalbumin; RAG: regeneration-associated gene; ROS: reactive oxygen species; SARM1: sterile alpha and HEAT/Armadillo motif containing 1; SYN1: synapsin I.


Subject(s)
Calcitonin , Charcot-Marie-Tooth Disease , Armadillo Domain Proteins/genetics , Autophagy , Axons , Cytoskeletal Proteins/genetics , Reactive Oxygen Species , Animals , Mice
7.
Nephron ; 148(4): 230-244, 2024.
Article in English | MEDLINE | ID: mdl-37956656

ABSTRACT

INTRODUCTION: Suppressor of cytokine signaling 3 (SOCS3) is highly expressed in mice with renal ischemia/reperfusion (RI/R) injury and has the potential to regulate mitophagy. On this basis, this study further investigates the possible mechanism via which SOCS3 affects RI/R by regulating mitophagy. METHOD: After establishing a RI/R injury mouse model and a hypoxia/reoxygenation (H/R) cell model, the effects of silenced SOCS3 on injury and mitophagy in the above models were analyzed by ELISA, quantitative real-time polymerase chain reaction, Western blot, pathological sections, CCK-8 assay, flow cytometry, and JC-1 assay. Mechanistic studies were carried out with the help of database analysis and binding validation experiments (chromatin immunoprecipitation, dual-luciferase reporter assay, and co-immunoprecipitation). After the binding target was identified, the regulatory relationship between the target gene and SOCS3 was verified by rescue experiments. RESULT: The large increase in blood urea nitrogen (BUN) and creatinine (Cr) levels verified the success of the RI/R model. SOCS3 expression was up-regulated in RI/R mice. Silenced SOCS3 alleviated kidney damage and mitochondrial abnormalities in RI/R mice and inhibited mitophagy at the molecular level. Likewise, silenced SOCS3 alleviated H/R-induced cell damage and mitophagy. Finally, activating transcription factor 3 (ATF3) was determined to bind to the promoter of SOCS3, which interacted with insulin-like growth factor 1 receptor (IGF1R). Rescue experiments confirmed the effect of ATF3 on SOCS3 expression and the underlying regulatory mechanism. CONCLUSION: ATF3 mediates SOCS3 expression to promote the activation of mitophagy, thereby aggravating renal ischemia-reperfusion injury.


Subject(s)
Kidney Diseases , Reperfusion Injury , Animals , Mice , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 3/pharmacology , Gene Expression Regulation , Kidney/pathology , Kidney Diseases/pathology , Mitophagy , Reperfusion Injury/metabolism
8.
Int J Mol Sci ; 24(13)2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37446172

ABSTRACT

Metformin is one of the most commonly used drugs for type 2 diabetes mellitus. In addition to its anti-diabetic property, evidence suggests more potential applications for metformin, such as antiaging, cellular protection, and anti-inflammation. Studies have reported that metformin activates pathways with anti-inflammatory effects, enhances the integrity of gut epithelial tight junctions, and promotes a healthy gut microbiome. These actions contribute to the protective effect of metformin against gastrointestinal (GI) tract injury. However, whether metformin plays a protective role in psychological-stress-associated GI tract injury remains elusive. We aim to elucidate the potential protective effect of metformin on the GI system and develop an effective intervention strategy to counteract GI injury induced by acute psychological stress. By monitoring the levels of GI-nonabsorbable Evans blue dye in the bloodstream, we assessed the progression of GI injury in live mice. Our findings demonstrate that the administration of metformin effectively mitigated GI leakage caused by psychological stress. The GI protective effect of metformin is more potent when used on wild-type mice than on activating-transcription-factor 3 (ATF3)-deficient (ATF3-/-) mice. As such, metformin-mediated rescue was conducted in an ATF3-dependent manner. In addition, metformin-mediated protection is associated with the induction of stress-induced GI mRNA expressions of the stress-induced genes ATF3 and AMP-activated protein kinase. Furthermore, metformin treatment-mediated protection of CD326+ GI epithelial cells against stress-induced apoptotic cell death was observed in wild-type but not in ATF3-/- mice. These results suggest that metformin plays a protective role in stress-induced GI injury and that ATF3 is an essential regulator for metformin-mediated rescue of stress-induced GI tract injury.


Subject(s)
Diabetes Mellitus, Type 2 , Metformin , Mice , Animals , Activating Transcription Factor 3/genetics , Metformin/pharmacology , Epithelial Cells/metabolism , AMP-Activated Protein Kinases
9.
Clin Respir J ; 2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37385291

ABSTRACT

INTRODUCTION: Bronchial asthma (BA) is a heterogeneous disease characterized by chronic airway inflammation. This study investigated the serum miR-27a-3p/activating transcription factor 3 (ATF3) expression in children with BA and their correlations with airway inflammation. METHODS: Children with BA (N = 120) and healthy children (N = 108) were enrolled. Serum levels of interleukin (IL)-17, IL-6, tumor necrosis factor (TNF)-α, immunoglobulin E (IgE), miR-27a-3p, ATF3, and the number of eosinophils (EOS) were measured using enzyme-linked immunosorbent assay (ELISA), reverse transcription quantitative polymerase chain reaction (RT-qPCR), and an automatic hematology analyzer. The correlations between miR-27a-3p and ATF3 and between miR-27a-3p/ATF3 and inflammation-related factors were analyzed by the Pearson method. The diagnostic values of miR-27a-3p and ATF3 in BA were evaluated using receiver operating characteristic (ROC) curves. The influencing factors of BA were assessed using multivariate logistic regression. Finally, the targeting relation between miR-27a-3p and ATF3 was predicted and analyzed by TargetScan and Starbase databases, and dual-luciferase assay. RESULTS: There were significant differences in forced expiratory volume in 1 s (FEV1)% predicted and FEV1/forced vital capacity (FVC)%, serum levels of IgE, IL-17, IL-6, and TNF-α, and EOS numbers between healthy children and BA children. Serum miR-27a-3p was negatively correlated with ATF3 and positively correlated with inflammation-related factors in BA children. Serum ATF3 mRNA levels were negatively correlated with inflammatory factors in BA children. miR-27a-3p and ATF3 had good diagnostic values in BA children. FEV% predicted, IL-6, TNF-α, miR-27a-3p, and ATF3 were independent risk factors for BA. miR-27a-3p targeted ATF3. CONCLUSION: Serum miR-27a-3p was highly expressed, whereas ATF3 was poorly expressed in BA children, and they were significantly correlated with airway inflammation, had good diagnostic values in BA children, and were independent risk factors for asthma.

10.
Biomedicines ; 11(6)2023 May 23.
Article in English | MEDLINE | ID: mdl-37371606

ABSTRACT

Obesity is an emerging concern globally with increasing prevalence. Obesity is associated with many diseases, such as cardiovascular disease, dyslipidemia, and cancer. Thus, effective new antiobesity drugs should be urgently developed. We synthesized SW20.1, a compound that induces activating transcription factor 3 (ATF3) expression. The results of Oil Red O staining and quantitative real-time polymerase chain reaction revealed that SW20.1 was more effective in reducing lipid accumulation in 3T3-L1 preadipocytes than the previously synthesized ST32db, and that it inhibited the expression of the genes involved in adipogenesis and lipogenesis. A chromatin immunoprecipitation assay indicated that SW20.1 inhibited adipogenesis and lipogenesis by binding to the upstream promoter region of resistin at two sites (-2861/-2854 and -241/-234). In mice, the intraperitoneal administration of SW20.1 reduced body weight, white adipocyte weight in different regions, serum cholesterol levels, adipogenesis-related gene expression, hepatic steatosis, and serum resistin levels. Overall, SW20.1 exerts antiobesity effects by inhibiting resistin through the ATF3 pathway. Our study results indicate that SW20.1 is a promising therapeutic drug for diet-induced obesity.

11.
J Cereb Blood Flow Metab ; 43(2_suppl): 8-17, 2023 11.
Article in English | MEDLINE | ID: mdl-37165649

ABSTRACT

Activating transcription factor 3 (ATF3) is one of the most important transcription factors that respond to and exert dual effects on inflammatory responses. Recently, the involvement of ATF3 in the neuroinflammatory response to acute brain injury (ABI) has been highlighted. It functions by regulating neuroimmune activation and the production of neuroinflammatory mediators. Notably, recent clinical evidence suggests that ATF3 may serve as a potential ideal biomarker of the long-term prognosis of ABI patients. This mini-review describes the essential inflammation modulatory roles of ATF3 in different disease contexts and summarizes the regulatory mechanisms of ATF3 in the ABI-induced neuroinflammation.


Subject(s)
Activating Transcription Factor 3 , Brain Injuries , Mice , Animals , Humans , Activating Transcription Factor 3/metabolism , Neuroinflammatory Diseases , Mice, Knockout , Inflammation/metabolism
12.
J Pers Med ; 13(4)2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37109086

ABSTRACT

Understanding the regulatory mechanisms underlying corneal epithelial cell (CEC) proliferation in vitro may provide the means to boost CEC production in cell therapy for ocular disorders. The transcription factor ΔNp63 plays a crucial role in the proliferation of CECs, but the underlying mechanisms is yet to be elucidated. TP63 and ΔNp63 are encoded by the TP63 gene via alternative promoters. We previously reported that both ΔNp63 and activating transcription factor (ATF3) are substantially expressed in cultured CECs, but the regulatory relationship between ΔNp63 and ATF3 is unknown. In the present study, we found that ΔNp63 increased ATF3 expression and ATF3 promoter activity in cultured CECs. The deletion of the p63 binding core site reduced ATF3 promoter activity. CECs overexpressing ATF3 exhibited significantly greater proliferation than control CECs. ATF3 knockdown suppressed the ΔNp63-induced increase in cell proliferation. Overexpression of ATF3 in CECs significantly elevated protein and mRNA levels of cyclin D. The protein levels of keratin 3/14, integrin ß1, and involucrin did not differ between ATF3-overexpressing CECs, ATF3-downregulated CECs, and control cells. In conclusion, our results suggest that ΔNp63 increases CEC proliferation via the ΔNp63/ATF3/CDK pathway.

13.
Front Mol Neurosci ; 16: 1150296, 2023.
Article in English | MEDLINE | ID: mdl-37033378

ABSTRACT

Activating Transcription Factor 3 (ATF3) is upregulated in reaction to several cellular stressors found in a wide range of pathological conditions to coordinate a transcriptional response. ATF3 was first implicated in the transcriptional reaction to axotomy when its massive upregulation was measured in sensory and motor neuron cell bodies following peripheral nerve injury. It has since been shown to be critical for successful axon regeneration in the peripheral nervous system and a promising target to mitigate regenerative failure in the central nervous system. However, much of the research to date has focused on ATF3's function in neurons, leaving the expression, function, and therapeutic potential of ATF3 in glia largely unexplored. In the immunology literature ATF3 is seen as a master regulator of the innate immune system. Specifically, in macrophages following pathogen or damage associated molecular pattern receptor activation and subsequent cytokine release, ATF3 upregulation abrogates the inflammatory response. Importantly, ATF3 upregulation is not exclusively due to cellular stress exposure but has been achieved by the administration of several small molecules. In the central nervous system, microglia represent the resident macrophage population and are therefore of immediate interest with respect to ATF3 induction. It is our perspective that the potential of inducing ATF3 expression to dampen inflammatory microglial phenotype represents an unexplored therapeutic target and may have synergistic benefits when paired with concomitant neuronal ATF3 upregulation. This would be of particular benefit in pathologies that involve both detrimental inflammation and neuronal damage including spinal cord injury, multiple sclerosis, and stroke.

14.
Curr Mol Med ; 23(9): 981-990, 2023.
Article in English | MEDLINE | ID: mdl-37073154

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is a degenerative joint disease involving both cartilage and synovium. Activating transcription factor 3 (ATF3) and regulator of G protein signaling 1 (RGS1) have been reported to be up-regulated in OA. However, little is known regarding the relationship between these two genes and the mechanism of this relationship in OA development. Therefore, the present study explores the mechanism of ATF3-mediated RGS1 in the proliferation, migration, and apoptosis of synovial fibroblasts. METHODS: After the OA cell model was constructed with TGF-ß1 induction, human fibroblast-like synoviocytes (HFLSs) were transfected with ATF3 shRNA or RGS1 shRNA alone or co-transfected with ATF3 shRNA and pcDNA3.1-RGS1. Then, proliferation, migration, apoptosis, and the expression of ATF3, RGS1, α-SMA, BCL-2, caspase3, and cleaved-caspase3 were measured. Meanwhile, the potential relationship between ATF3 and RGS1 was predicted and validated. RESULTS AND DISCUSSION: Analysis of the GSE185059 dataset suggested that RGS1 was up-regulated in OA synovial fluid exosomes. Moreover, ATF3 and RGS1 were both highly expressed in TGF-ß1-induced HFLSs. Transfection of ATF3 shRNA or RGS1 shRNA significantly reduced proliferation and migration and promoted apoptosis of TGF- ß1-induced HFLSs. Mechanistically, ATF3 bound to the RGS1 promoter and elevated RGS1 expression. Silencing ATF3 repressed proliferation and migration and enhanced apoptosis of TGF-ß1-induced HFLSs by down-regulating RGS1. CONCLUSION: ATF3 binds to the RGS1 promoter and enhances RGS1 expression to accelerate cell proliferation and block cell apoptosis in TGF-ß1-induced synovial fibroblasts.


Subject(s)
RGS Proteins , Transforming Growth Factor beta1 , Humans , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Arthroscopy , Fibroblasts/metabolism , Apoptosis/genetics , Cell Proliferation/genetics , RNA, Small Interfering/metabolism , Cells, Cultured , RGS Proteins/genetics , RGS Proteins/metabolism
15.
Naunyn Schmiedebergs Arch Pharmacol ; 396(6): 1155-1169, 2023 06.
Article in English | MEDLINE | ID: mdl-36688958

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease that is characterized by abnormal proliferation of fibroblasts and extracellular matrix remodeling, ultimately leading to respiratory insufficiency or even death. Naringin (Nar), a natural compound derived from grapefruit and citrus fruits, has several pharmacological activities that are associated with therapeutic benefits for IPF. However, the specific molecular mechanisms underlying its pulmonary tissue-protective effects remain largely unknown. This study aimed to investigate the effects of Nar on endoplasmic reticulum stress (ERS) and mitophagy. A bleomycin (BLM)-induced mouse model of IPF was established for treatment with different doses of Nar. Histopathological changes in the lung were examined by hematoxylin and eosin (HE) staining and Masson staining. The extent of fibrosis was determined by measuring hydroxyproline and collagen expression levels. The levels of inflammatory cytokines and oxidative stress indicators were determined by Enzyme linked immunosorbent assay (ELISA) and biochemical kits. Western blot and immunofluorescence were used to evaluate the expression levels of the mitophagy-related markers. Cell apoptosis was estimated by western blot and TUNEL staining. Nar reduced the levels of inflammatory response, oxidative stress and decreased the proportion of apoptosis. Nar also inhibited the expression of the ERS and mitophagy-related genes and ERS-downstream proteins, thereby activating transcription factor (ATF) 3 and inhibiting the transcription of PTEN-induced kinase 1 (PINK1). Taken together, Nar is a promising therapeutic agent for treating IPF via inhibiting ERS, reducing apoptosis, and maintaining mitochondrial homeostasis, all of which may be associated with the regulation of the ATF3/PINK1 signaling axis.


Subject(s)
Pulmonary Fibrosis , Mice , Animals , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Mitophagy , Protein Kinases/metabolism , Endoplasmic Reticulum Stress , Bleomycin/adverse effects
16.
Eur J Med Chem ; 246: 114951, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36455354

ABSTRACT

The induction of activating transcription factor 3 (ATF3) was identified as a promising therapeutic mechanism to overcome metabolic syndrome. Hence, a structure-activity relationship campaign on the chiral lead (1b) was conducted with a scaffold-hopping approach, whereby achiral 7-methoxy-3-methyl-1H-chromeno[4,3-c]pyrazol-4-one (16c) was recognized as a potential ATF3 inducer with a lipid-lowering feature in a pre-differentiated 3T3-L1 cell model. Also, in a high-fat diet scenario, mice subjected to 16c demonstrated robust weight loss with shrinkage of the white adipose mass and fewer hypertrophic adipocytes, accompanied by a preferable glycemic profile compared to 1b. Additionally, the biochemical analysis revealed that 16c further ameliorated the liver function and improved the plasma triglyceride profile that were absent from mice treated with 1b. Taken together, 16c shows promise as an ATF3 stimulant for further development to alleviate metabolic syndrome.


Subject(s)
Metabolic Syndrome , Mice , Animals , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Activating Transcription Factor 3/metabolism , Obesity/metabolism , Adipocytes/metabolism , Cell Differentiation , 3T3-L1 Cells , Diet, High-Fat/adverse effects
17.
Neurobiol Stress ; 21: 100495, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36532375

ABSTRACT

Stress-induced neuroinflammation is considered an important mechanism in the pathogenesis of depression. As immune effector cells in the brain, microglia play an essential role in neuroinflammation under stress, but the underlying mechanism remains controversial. Here, we performed RNA-seq and ATAC-seq to study microglia-specific epigenomic changes in mice after 12 weeks of exposure to mild stress. Our study revealed that chronic stress induced pronounced anxiety and depressive-like behavioral changes. However, microglia did not manifest a state of neuroinflammatory activation; instead, they displayed morphological changes characterized by hyper-ramification. Furthermore, we revealed large-scale transcriptional repression in microglia isolated from the stressed brain, including many interferon (IFN)-regulated genes (IRGs) and some encompassing DNA repeats. GSEA showed that the down-regulated genes were enriched in the IFN-mediated neuroimmune signaling pathways. In addition, integrative analysis with a published scRNA-seq dataset revealed that these down-regulated genes were enriched in a distinct subpopulation of "Interferon microglia". ATAC-seq analysis further showed that differential gene expression was positively correlated with the changes in chromatin accessibility, and the IFN-stimulated response element (ISRE) was enriched in the down-regulated ATAC-seq loci. Interestingly, this phenotype was not associated with the production of IFNs. Instead, the gene encoding Activating Transcription Factor 3 (ATF3) was significantly increased in the stressed microglia, which might contribute to the transcriptional repression of IRGs. Our study reported microglia-specific transcriptional repression of IRGs independent of the production of IFNs, providing some new insights into neuroimmune dysregulation under prolonged stress.

18.
Mol Med Rep ; 26(5)2022 Nov.
Article in English | MEDLINE | ID: mdl-36102309

ABSTRACT

Depletion of activating transcription factor 3 (ATF3) expression has previously been reported to promote hypertrophy, dysfunction and fibrosis in stress overload­induced hearts; however, the mechanism involved remains poorly understood. In the present study, the mechanism underlying the activation of cysteine­rich angiogenic protein 61 (Cyr61) by ATF3 in hyperproliferative and fibrotic human cardiac fibroblasts (HCFs), induced by angiotensin II (Ang II), was evaluated. The mRNA and protein expression levels of ATF3 and Cyr61 were assessed using reverse transcription­quantitative PCR and western blotting, respectively. The Cell Counting Kit­8 assay was used to assess cell viability. Cell migration was assessed using the wound healing assay and western blotting, whereas the extent of cell fibrosis was evaluated using immunofluorescence staining and western blotting. The binding site of ATF3 to the Cyr61 promoter was predicted using the JASPAR database, and verified using luciferase reporter and chromatin immunoprecipitation assays. The results demonstrated that the mRNA and protein expression levels of ATF3 were significantly upregulated in Ang II­induced HCFs. Overexpression of ATF3 significantly inhibited the Ang II­induced viability, migration and fibrosis of HCFs, whereas ATF3 knockdown mediated significant opposing effects. Mechanistically, ATF3 was demonstrated to transcriptionally activate Cyr61. Cyr61 silencing was subsequently revealed to reverse the effects of ATF3 overexpression on HCFs potentially via regulation of the TGF­ß/Smad signaling pathway. The results of the present study suggested that ATF3 could suppress HCF viability and fibrosis via the TGF­ß/Smad signaling pathway by activating the transcription of Cyr61.


Subject(s)
Activating Transcription Factor 3/metabolism , Angiotensin II , Cysteine-Rich Protein 61/metabolism , Activating Transcription Factor 3/genetics , Angiogenic Proteins , Angiotensin II/pharmacology , Cyclic AMP Response Element-Binding Protein , Cysteine , Fibrosis , Humans , Myocytes, Cardiac/metabolism , RNA, Messenger/metabolism , Transforming Growth Factor beta/metabolism
19.
Cancer Lett ; 550: 215919, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36116741

ABSTRACT

In cancer cells, poly (ADP-ribose) polymerase (PARP)-1 and PARP2 initiate and regulate DNA repair pathways to protect against DNA damage and cell death caused by radiotherapy or chemotherapy. Radiotherapy and PARP inhibitors (PARPis) have been combined in clinical trials, but their action mechanisms remain unclear. Here, we show that activated by ionizing radiation (IR) generated dsDNA, cyclic GMP-AMP synthase (cGAS) signaling promoted regulated cell death, specifically ferroptosis, via the activating transcription factor 3 (ATF3)-solute carrier family 7 member 11 axis and the antitumor immune response via the interferon-ß-CD8+ T cell pathway. Niraparib, a widely used PARPi, augmented cGAS-mediated ferroptosis and immune activation. In colorectal cancer models, cGAS knockdown (KD) compromised IR-induced ferroptosis via downregulation of ATF3 (key ferroptosis regulator) expression. cGAS depletion reversed IR-induced infiltration of CD8+ T or CD8+GZMB+ T cells in the cGAS KD group. Survival analysis of paired tumor samples before and after standard radiotherapy revealed that high expression levels of cGAS, ATF3, and PTGS2 and high density of CD8+ T cells resulted in a significantly high disease-free survival rate in patients with rectal cancer. Therefore, PARPi treatment increases the cytoplasmic accumulation of dsDNA caused by IR, triggering the cGAS signaling-mediated tumor control in cancer cell lines and mouse xenograft models.


Subject(s)
Colorectal Neoplasms , Ferroptosis , Activating Transcription Factor 3 , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Animals , CD8-Positive T-Lymphocytes , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Cyclooxygenase 2/metabolism , Humans , Immunity , Interferon-beta/pharmacology , Membrane Proteins/metabolism , Mice , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Ribose/metabolism , Ribose/pharmacology , Signal Transduction
20.
Nutrients ; 14(18)2022 Sep 12.
Article in English | MEDLINE | ID: mdl-36145135

ABSTRACT

High fructose intake has been implicated in obesity and metabolic syndrome, which are related to increased cardiovascular mortality. However, few studies have experimentally examined the role of renin-angiotensin system blockers and calcium channel blockers (CCB) in obesity. We investigated the effects of valsartan (an angiotensin II receptor blocker) and amlodipine (a CCB) on lipolysis through the potential mechanism of PU.1 inhibition. We observed that high fructose concentrations significantly increased adipose size and triglyceride, monoacylglycerol lipase, adipose triglyceride lipase, and stearoyl-CoA desaturase-1 (SCD1), activating transcription factor 3 and PU.1 levels in adipocytes in vitro. Subsequently, PU.1 inhibitor treatment was able to reduce triglyceride, SCD1, and PU.1 levels. In addition, elevated levels of triglyceride and PU.1, stimulated by a high fructose concentration, decreased with valsartan and amlodipine treatment. Overall, these findings suggest that high fructose concentrations cause triacylglycerol storage in adipocytes through PU.1-mediated activation. Furthermore, valsartan and amlodipine treatment reduced triacylglycerol storage in adipocytes by inhibiting PU.1 activation in high fructose concentrations in vitro. Thus, the benefits of valsartan and amlodipine in lipolysis may be through PU.1 inhibition in fructose-induced adiposity, and PU.1 inhibition might have a potential therapeutic role in lipolysis in fructose-induced obesity.


Subject(s)
Amlodipine , Hypertension , Activating Transcription Factor 3/metabolism , Adiposity , Amlodipine/pharmacology , Angiotensin Receptor Antagonists , Antihypertensive Agents/pharmacology , Blood Pressure , Calcium Channel Blockers/pharmacology , Fructose/adverse effects , Humans , Hypertension/drug therapy , Lipase/metabolism , Lipolysis , Monoacylglycerol Lipases/metabolism , Obesity/drug therapy , Obesity/etiology , Stearoyl-CoA Desaturase/metabolism , Tetrazoles/pharmacology , Tetrazoles/therapeutic use , Triglycerides/pharmacology , Valsartan/metabolism , Valsartan/pharmacology , Valsartan/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...