Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.322
Filter
1.
Am J Physiol Cell Physiol ; 327(4): C901-C912, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39129491

ABSTRACT

Pulmonary arterial hypertension (PAH) is a debilitating vascular disorder characterized by abnormal pulmonary artery smooth muscle cell (PASMC) proliferation and collagen synthesis, contributing to vascular remodeling and elevated pulmonary vascular resistance. This study investigated the critical role of 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inosine monophosphate cyclohydrolase (ATIC) in cell proliferation and collagen synthesis in PASMCs in PAH. Here we show that ATIC levels are significantly increased in the lungs of monocrotaline (MCT)-induced PAH rat model, hypoxia-induced PAH mouse model, and platelet-derived growth factor (PDGF)-stimulated PASMCs. Inhibition of ATIC attenuated PDGF-induced cell proliferation and collagen I synthesis in PASMCs. Conversely, overexpression or knockdown of ATIC causes a significant promotion or inhibition of Ras and ERK activation, cell proliferation, and collagen synthesis in PASMCs. Moreover, ATIC deficiency attenuated Ras activation in the lungs of hypoxia-induced PAH mice. Furthermore, Ras inhibition attenuates ATIC overexpression- and PDGF-induced collagen synthesis and PASMC proliferation. Notably, we identified that transcription factors MYC, early growth response protein 1 (EGR1), and specificity protein 1 (SP1) directly binds to promoters of Atic gene and regulate ATIC expression. These results provide the first evidence that ATIC promotes PASMC proliferation in pulmonary vascular remodeling through the Ras signaling pathway.NEW & NOTEWORTHY Our findings highlight the important role of ATIC in the PASMC proliferation of pulmonary vascular remodeling through its modulation of the Ras signaling pathway and its regulation by transcription factors MYC, EGR1, and SP1. ATIC's modulation of Ras signal pathway represents a novel mechanism contributing to PAH development.


Subject(s)
Cell Proliferation , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Pulmonary Artery , Rats, Sprague-Dawley , Signal Transduction , Animals , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/drug effects , Rats , Male , Mice , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , ras Proteins/metabolism , ras Proteins/genetics , Hydroxymethyl and Formyl Transferases/metabolism , Hydroxymethyl and Formyl Transferases/genetics , Mice, Inbred C57BL , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/enzymology , Platelet-Derived Growth Factor/metabolism , Disease Models, Animal , Cells, Cultured , Vascular Remodeling/drug effects , Monocrotaline/toxicity
2.
Oral Oncol ; 158: 106999, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39197193

ABSTRACT

Regulatory B (Breg) cells is a type of immune cell that exhibit immunosuppressive behavior within the tumor microenvironment. However, the differentiation and regulatory mechanisms of these Breg cells remain unexplored. Single-cell transcriptome sequencing analysis of human nasopharyngeal carcinoma (NPC) revealed a significant enrichment of B cell subset characterized by high expression of EGR1 and EGR3 in the tumor microenvironment. Notably, in the hypoxic microenvironment, these B cells induce MAPK pathway activation, subsequently triggering the activation of transcription factors EGR1 and EGR3, which further modulate the expression of immunosuppressive factors like TGFB1 and IL10. In transplant experiments using primary B cells induced under hypoxia and co-transplanted with cancer cells, a significant increase in tumor growth was observed. Mechanism experiments demonstrated that EGR1hi and EGR3+ B cells further activate the maturation and immunosuppressive function of Treg cells through the secretion of IL16 and TNF-α. Hence, this study identifies the key transcription factors EGR1 and EGR3 as essential regulators and elucidates the differentiation of Breg cells under hypoxic conditions.


Subject(s)
Early Growth Response Protein 1 , Early Growth Response Protein 3 , Nasopharyngeal Carcinoma , Tumor Microenvironment , Humans , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Carcinoma/genetics , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 3/metabolism , Early Growth Response Protein 3/genetics , Animals , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/genetics , Mice , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Cell Line, Tumor , Cell Hypoxia
3.
Nat Cardiovasc Res ; 3(8): 915-932, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39196027

ABSTRACT

Senescence plays a key role in various physiological and pathological processes. We reported that injury-induced transient senescence correlates with heart regeneration, yet the multi-omics profile and molecular underpinnings of regenerative senescence remain obscure. Using proteomics and single-cell RNA sequencing, here we report the regenerative senescence multi-omic signature in the adult mouse heart and establish its role in neonatal heart regeneration and agrin-mediated cardiac repair in adult mice. We identified early growth response protein 1 (Egr1) as a regulator of regenerative senescence in both models. In the neonatal heart, Egr1 facilitates angiogenesis and cardiomyocyte proliferation. In adult hearts, agrin-induced senescence and repair require Egr1, activated by the integrin-FAK-ERK-Akt1 axis in cardiac fibroblasts. We also identified cathepsins as injury-induced senescence-associated secretory phenotype components that promote extracellular matrix degradation and potentially assist in reducing fibrosis. Altogether, we uncovered the molecular signature and functional benefits of regenerative senescence during heart regeneration, with Egr1 orchestrating the process.


Subject(s)
Cell Proliferation , Cellular Senescence , Early Growth Response Protein 1 , Myocytes, Cardiac , Regeneration , Animals , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Regeneration/physiology , Cellular Senescence/physiology , Myocytes, Cardiac/metabolism , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Signal Transduction , Fibroblasts/metabolism , Mice , Proto-Oncogene Proteins c-akt/metabolism , Cells, Cultured , Animals, Newborn , Disease Models, Animal , Senescence-Associated Secretory Phenotype , Proteomics , Single-Cell Analysis , Male , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice, Knockout , Focal Adhesion Kinase 1
4.
Sci Adv ; 10(31): eadk8232, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093963

ABSTRACT

While extracellular matrix (ECM) stress relaxation is increasingly appreciated to regulate stem cell fate commitment and other behaviors, much remains unknown about how cells process stress-relaxation cues in tissue-like three-dimensional (3D) geometries versus traditional 2D cell culture. Here, we develop an oligonucleotide-crosslinked hyaluronic acid-based ECM platform with tunable stress relaxation properties capable of use in either 2D or 3D. Strikingly, stress relaxation favors neural stem cell (NSC) neurogenesis in 3D but suppresses it in 2D. RNA sequencing and functional studies implicate the membrane-associated protein spectrin as a key 3D-specific transducer of stress-relaxation cues. Confining stress drives spectrin's recruitment to the F-actin cytoskeleton, where it mechanically reinforces the cortex and potentiates mechanotransductive signaling. Increased spectrin expression is also accompanied by increased expression of the transcription factor EGR1, which we previously showed mediates NSC stiffness-dependent lineage commitment in 3D. Our work highlights spectrin as an important molecular sensor and transducer of 3D stress-relaxation cues.


Subject(s)
Cell Lineage , Extracellular Matrix , Neural Stem Cells , Spectrin , Spectrin/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Extracellular Matrix/metabolism , Animals , Mice , Cell Differentiation , Mechanotransduction, Cellular , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Neurogenesis , Actin Cytoskeleton/metabolism , Stress, Mechanical , Humans , Cell Culture Techniques/methods
5.
Tissue Cell ; 90: 102510, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39126833

ABSTRACT

The epithelial-mesenchymal transition (EMT) plays a crucial role in lung cancer metastasis, rendering it a promising therapeutic target. Research has shown that non-small cell lung cancer (NSCLC) with p53 mutations exhibits an increased tendency for cancer metastasis. However, the exact contribution of the p53-R273H mutation to tumor metastasis remains uncertain in the current literature. Our study established the H1299-p53-R273H cell model successfully by transfecting the p53-R273H plasmid into H1299 cells. We observed that p53-R273H promotes cell proliferation, migration, invasion, and EMT through CCK-8, wound healing, transwell, western blot and immunofluorescence assays. Notably, the expression of EGR1 was increased in H1299-p53-R273H cells. Knocking out EGR1 in these cells hindered the progression of EMT. ChIP-PCR experiments revealed that p53-R273H binds to the EGR1 promoter sequence, thereby regulating its expression. These findings suggest that p53-R273H triggers EMT by activating EGR1, thereby offering a potential therapeutic approach for lung cancer treatment.


Subject(s)
Cell Movement , Cell Proliferation , Early Growth Response Protein 1 , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Tumor Suppressor Protein p53 , Epithelial-Mesenchymal Transition/genetics , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Cell Movement/genetics , Cell Proliferation/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Mutation , Neoplasm Invasiveness/genetics
6.
J Physiol ; 602(17): 4171-4193, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39159314

ABSTRACT

The objective of this work was to investigate myonuclear permanence and transcriptional regulation as mechanisms for cellular muscle memory after strength training in humans. Twelve untrained men and women performed 10 weeks of unilateral elbow-flexor strength training followed by 16 weeks of de-training. Thereafter, 10 weeks' re-training was conducted with both arms: the previously trained arm and the contralateral untrained control arm. Muscle biopsies were taken from the trained arm before and after both training periods and from the control arm before and after re-training. Muscle biopsies were analysed for fibre cross-sectional area (fCSA), myonuclei and global transcriptomics (RNA sequencing). During the first training period, myonuclei increased in type 1 (13 ± 17%) and type 2 (33 ± 23%) fibres together with a 30 ± 43% non-significant increase in mixed fibre fCSA (P = 0.069). Following de-training, fCSA decreased in both fibre types, whereas myonuclei were maintained, resulting in 33% higher myonuclear number in previously trained vs. control muscle in type 2 fibres. Furthermore, in the previously trained muscle, three differentially expressed genes (DEGs; EGR1, MYL5 and COL1A1) were observed. Following re-training, the previously trained muscle showed larger type 2 fCSA compared to the control (P = 0.035). However, delta change in type 2 fCSA was not different between muscles. Gene expression was more dramatically changed in the control arm (1338 DEGs) than in the previously trained arm (822 DEGs). The sustained higher number of myonuclei in the previously trained muscle confirms myonuclear accretion and permanence in humans. Nevertheless, because of the unclear effect on the subsequent hypertrophy with re-training, the physiological benefit remains to be determined. KEY POINTS: Muscle memory is a cellular mechanism that describes the capacity of skeletal muscle fibres to respond differently to training stimuli if the stimuli have been previously encountered. This study overcomes past methodological limitations related to the choice of muscles and analytical procedures. We show that myonuclear number is increased after strength training and maintained during de-training. Increased myonuclear number and differentially expressed genes related to muscle performance and development in the previously trained muscle did not translate into a clearly superior responses during re-training. Because of the unclear effect on the subsequent hypertrophy and muscle strength gain with re-training, the physiological benefit remains to be determined.


Subject(s)
Resistance Training , Humans , Resistance Training/methods , Male , Female , Adult , Muscle, Skeletal/physiology , Muscle, Skeletal/metabolism , Young Adult , Gene Expression Regulation , Cell Nucleus/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Muscle Fibers, Skeletal/physiology , Muscle Fibers, Skeletal/metabolism , Transcription, Genetic , Transcriptome
7.
J Exp Clin Cancer Res ; 43(1): 204, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39044249

ABSTRACT

Endometrial cancer (EC) stem cells (ECSCs) are pivotal in the oncogenesis, metastasis, immune escape, chemoresistance, and recurrence of EC. However, the specific mechanism of stem cell maintenance in EC cells (ECCs) has not been clarified. We found that WTAP and m6A levels decreased in both EC and ECSCs, and that knocking down WTAP promoted ECCs and ECSCs properties, including proliferation, invasion, migration, cisplatin resistance, and self-renewal. The downregulation of WTAP leads to a decrease in the m6A modification of EGR1 mRNA, and it is difficult for IGF2BP3, as an m6A reader, to recognize and bind to EGR1 mRNA that has lost m6A modification, resulting in a decrease in the stability of EGR1 mRNA. A decrease in the EGR1 level led to a decrease of in the expression tumor suppressor gene PTEN, resulting in deregulation and loss of cellular homeostasis and thereby fostering EC stem cell traits. Notably, the enforced overexpression of WTAP, EGR1, and PTEN inhibited the oncogenic effects of ECCs and ECSCs in vivo, and the combined overexpression of WTAP + EGR1 and EGR1 + PTEN further diminished the tumorigenic potential of these cells. Our findings revealed that the WTAP/EGR1/PTEN pathway is important regulator of EC stem cell maintenance, chemotherapeutic resistance, and tumorigenesis, suggesting a novel and promising therapeutic avenue for treating EC.


Subject(s)
Early Growth Response Protein 1 , Endometrial Neoplasms , Neoplastic Stem Cells , PTEN Phosphohydrolase , RNA-Binding Proteins , Humans , Female , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Endometrial Neoplasms/genetics , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Mice , Animals , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Line, Tumor , Phenotype , Cell Proliferation , Gene Expression Regulation, Neoplastic , Adenosine/analogs & derivatives , Adenosine/metabolism
8.
J Am Chem Soc ; 146(30): 20685-20699, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39012486

ABSTRACT

The primer-guided entropy-driven high-throughput evolution of the DNA-based constitutional dynamic network, CDN, is introduced. The entropy gain associated with the process provides a catalytic principle for the amplified emergence of the CDN. The concept is applied to develop a programmable, spatially localized DNA circuit for effective in vitro and in vivo theranostic, gene-regulated treatment of cancer cells. The localized circuit consists of a DNA tetrahedron core modified at its corners with four tethers that include encoded base sequences exhibiting the capacity to emerge and assemble into a [2 × 2] CDN. Two of the tethers are caged by a pair of siRNA subunits, blocking the circuit into a mute, dynamically inactive configuration. In the presence of miRNA-21 as primer, the siRNA subunits are displaced, resulting in amplified release of the siRNAs silencing the HIF-1α mRNA and fast dynamic reconfiguration of the tethers into a CDN. The resulting CDN is, however, engineered to be dynamically reconfigured by miRNA-155 into an equilibrated mixture enriched with a DNAzyme component, catalyzing the cleavage of EGR-1 mRNA. The DNA tetrahedron nanostructure stimulates enhanced permeation into cancer cells. The miRNA-triggered entropy-driven reconfiguration of the spatially localized circuit leads to the programmable, cooperative bis-gene-silencing of HIF-1α and EGR-1 mRNAs, resulting in the effective and selective apoptosis of breast cancer cells and effective inhibition of tumors in tumor bearing mice.


Subject(s)
DNA , Entropy , Genetic Therapy , MicroRNAs , Humans , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , MicroRNAs/chemistry , DNA/chemistry , Mice , RNA, Small Interfering/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cell Line, Tumor , Breast Neoplasms/drug therapy , Female , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , DNA, Catalytic/genetics
9.
J Neurovirol ; 30(3): 286-302, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38926255

ABSTRACT

Caffeine is one of the most popular consumed psychostimulants that mitigates several neurodegenerative diseases. Nevertheless, the roles and molecular mechanisms of caffeine in HIV-associated neurocognitive disorders (HAND) remain largely unclear. Transactivator of transcription (Tat) is a major contributor to the neuropathogenesis of HAND in the central nervous system. In the present study, we determined that caffeine (100 µM) treatment significantly ameliorated Tat-induced decreased astrocytic viability, oxidative stress, inflammatory response and excessive glutamate and ATP release, thereby protecting neurons from apoptosis. Subsequently, SIRT3 was demonstrated to display neuroprotective effects against Tat during caffeine treatment. In addition, Tat downregulated SIRT3 expression via activation of EGR1 signaling, which was reversed by caffeine treatment in astrocytes. Overexpression of EGR1 entirely abolished the neuroprotective effects of caffeine against Tat. Furthermore, counteracting Tat or caffeine-induced differential expression of SIRT3 abrogated the neuroprotection of caffeine against Tat-triggered astrocytic dysfunction and neuronal apoptosis. Taken together, our study establishes that caffeine ameliorates astrocytes-mediated Tat neurotoxicity by targeting EGR1/SIRT3 signaling pathway. Our findings highlight the beneficial effects of caffeine on Tat-induced astrocytic dysfunction and neuronal death and propose that caffeine might be a novel therapeutic drug for relief of HAND.


Subject(s)
Apoptosis , Astrocytes , Caffeine , Early Growth Response Protein 1 , HIV-1 , Neurons , Signal Transduction , Sirtuin 3 , tat Gene Products, Human Immunodeficiency Virus , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/virology , Astrocytes/pathology , Sirtuin 3/genetics , Sirtuin 3/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Signal Transduction/drug effects , Caffeine/pharmacology , Humans , Apoptosis/drug effects , HIV-1/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurons/virology , Up-Regulation/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Cell Survival/drug effects , Cells, Cultured , AIDS Dementia Complex/drug therapy , AIDS Dementia Complex/genetics , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/pathology , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/toxicity
10.
J Cell Sci ; 137(12)2024 06 15.
Article in English | MEDLINE | ID: mdl-38940347

ABSTRACT

Some chemotherapy drugs modulate the formation of stress granules (SGs), which are RNA-containing cytoplasmic foci contributing to stress response pathways. How SGs mechanistically contribute to pro-survival or pro-apoptotic functions must be better defined. The chemotherapy drug lomustine promotes SG formation by activating the stress-sensing eIF2α kinase HRI (encoded by the EIF2AK1 gene). Here, we applied a DNA microarray-based transcriptome analysis to determine the genes modulated by lomustine-induced stress and suggest roles for SGs in this process. We found that the expression of the pro-apoptotic EGR1 gene was specifically regulated in cells upon lomustine treatment. The appearance of EGR1-encoding mRNA in SGs correlated with a decrease in EGR1 mRNA translation. Specifically, EGR1 mRNA was sequestered to SGs upon lomustine treatment, probably preventing its ribosome translation and consequently limiting the degree of apoptosis. Our data support the model where SGs can selectively sequester specific mRNAs in a stress-specific manner, modulate their availability for translation, and thus determine the fate of a stressed cell.


Subject(s)
Early Growth Response Protein 1 , Lomustine , RNA, Messenger , Humans , RNA, Messenger/metabolism , RNA, Messenger/genetics , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Lomustine/pharmacology , Stress Granules/metabolism , Stress Granules/genetics , Apoptosis/drug effects , Antineoplastic Agents, Alkylating/pharmacology
11.
Sci Rep ; 14(1): 14811, 2024 06 27.
Article in English | MEDLINE | ID: mdl-38926457

ABSTRACT

Ischemic heart diseases are a major global cause of death, and despite timely revascularization, heart failure due to ischemia-hypoxia reperfusion (IH/R) injury remains a concern. The study focused on the role of Early Growth Response 1 (EGR1) in IH/R-induced apoptosis in human cardiomyocytes (CMs). Human induced pluripotent stem cell (hiPSC)-derived CMs were cultured under IH/R conditions, revealing higher EGR1 expression in the IH/R group through quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting (WB). Immunofluorescence analysis (IFA) showed an increased ratio of cleaved Caspase-3-positive apoptotic cells in the IH/R group. Using siRNA for EGR1 successfully downregulated EGR1, suppressing cleaved Caspase-3-positive apoptotic cell ratio. Bioinformatic analysis indicated that EGR1 is a plausible target of miR-124-3p under IH/R conditions. The miR-124-3p mimic, predicted to antagonize EGR1 mRNA, downregulated EGR1 under IH/R conditions in qRT-PCR and WB, as confirmed by IFA. The suppression of EGR1 by the miR-124-3p mimic subsequently reduced CM apoptosis. The study suggests that treatment with miR-124-3p targeting EGR1 could be a potential novel therapeutic approach for cardioprotection in ischemic heart diseases in the future.


Subject(s)
Apoptosis , Down-Regulation , Early Growth Response Protein 1 , Induced Pluripotent Stem Cells , MicroRNAs , Myocytes, Cardiac , MicroRNAs/genetics , MicroRNAs/metabolism , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Humans , Myocytes, Cardiac/metabolism , Induced Pluripotent Stem Cells/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology
12.
Aging (Albany NY) ; 16(12): 10216-10238, 2024 06 28.
Article in English | MEDLINE | ID: mdl-38943627

ABSTRACT

This study aimed to reveal the specific role of early growth response protein 1 (EGR1) and nuclear receptor 4A3 (NR4A3) in nucleus pulposus cells (NPCs) and the related molecular mechanism and to identify a new strategy for treating intervertebral disc degeneration (IVDD). Bioinformatics analysis was used to explore and predict IVDD-related differentially expressed genes, and chromatin immunoprecipitation sequencing (ChIP-seq) revealed NR4A3 as the EGR1 target gene. An in vitro NPC model induced by tributyl hydrogen peroxide (TBHP) and a rat model induced by fibrous ring acupuncture were established. Western blotting, quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemical staining, immunofluorescence staining, and flow cytometry were used to detect the effects of EGR1 and NR4A3 knockdown and overexpression on NPC apoptosis and the expression of extracellular matrix (ECM) anabolism-related proteins. Interactions between EGR1 and NR4A3 were analyzed via ChIP-qPCR and dual luciferase assays. EGR1 and NR4A3 expression levels were significantly higher in severely degenerated discs (SDD) than in mildly degenerated discs (MDD), indicating that these genes are important risk factors in IVDD progression. ChIP-seq and RNA-seq revealed NR4A3 as a direct downstream target of EGR1, and this finding was verified by ChIP-qPCR and dual luciferase reporter experiments. Remarkably, the rescue experiments showed that EGR1 promotes TBHP-induced NPC apoptosis and impairs ECM anabolism, dependent on elevated NR4A3 expression. In summary, the EGR1-NR4A3 axis mediates the progression of NPC apoptosis and ECM impairment and is a potential therapeutic target in IVDD.


Subject(s)
Apoptosis , Early Growth Response Protein 1 , Intervertebral Disc Degeneration , Nucleus Pulposus , Oxidative Stress , Receptors, Thyroid Hormone , Adult , Animals , Female , Humans , Male , Middle Aged , Rats , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/pathology , Nerve Tissue Proteins , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , Rats, Sprague-Dawley , Receptors, Steroid/metabolism , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/metabolism , Receptors, Thyroid Hormone/genetics , Up-Regulation
13.
J Mol Histol ; 55(4): 437-454, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38874870

ABSTRACT

Anshen Shumai Decoction (ASSMD) is traditionally employed to manage coronary artery disease arrhythmias. Its protective efficacy against myocardial infarction remains to be elucidated. This investigation employed a rat model of myocardial infarction, achieved through the ligation of the left anterior descending (LAD) coronary artery, followed by a 28-day administration of ASSMD. The study observed the decoction's mitigative impact on myocardial injury, with gene regulation effects discerned through transcriptomic analysis. Furthermore, ASSMD's influence on cardiomyocyte apoptosis and fibrotic protein secretion was assessed using an embryonic rat cardiomyocyte cell line (H9c2) under hypoxic conditions and rat cardiac fibroblasts subjected to normoxic culture conditions with TGF-ß. A functional rescue assay involving overexpression of FOS and Early Growth Response Factor 1 (EGR1), combined with inhibition of the p38 Mitogen-activated Protein Kinase (MAPK) pathway, was conducted. Results indicated that ASSMD significantly curtailed cardiomyocyte apoptosis and myocardial fibrosis in infarcted rats, primarily by downregulating FOS and EGR1 gene expression and inhibiting the upstream p38 MAPK pathway. These actions of ASSMD culminated in reduced expression of pro-apoptotic, collagen, and fibrosis-associated proteins, conferring myocardial protection and anti-fibrotic effects on cardiac fibroblasts.


Subject(s)
Early Growth Response Protein 1 , Myocardial Infarction , Myocytes, Cardiac , Proto-Oncogene Proteins c-fos , p38 Mitogen-Activated Protein Kinases , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Rats , p38 Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Male , Apoptosis/drug effects , Ventricular Remodeling/drug effects , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Rats, Sprague-Dawley , Fibrosis , Cell Line , Signal Transduction/drug effects , Disease Models, Animal , MAP Kinase Signaling System/drug effects
14.
Antiviral Res ; 227: 105916, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38777095

ABSTRACT

The severe fever with thrombocytopenia syndrome virus (SFTSV) is a novel phlebovirus, recently being officially renamed as Dabie bandavirus, and a causative agent for an emerging infectious disease associated with high fatality. Effective therapeutics and vaccines are lacking and disease pathogenesis is yet to be fully elucidated. In our effort to identify new SFTSV inhibitory molecules, 6-Thioguanine (6-TG) was found to potently inhibit SFTSV infection. 6-TG has been widely used as therapeutic agent since the approval of the Food and Drug Administration in the 1960s. In the current study, we showed that 6-TG was a potent inhibitor of SFTSV infection with 50% effective concentrations (EC50) of 3.465 µM in VeroE6 cells, and 1.848 µM in HUVEC cells. The selectivity index (SI) was >57 in VeroE6 cells and >108 in HUVEC cells, respectively. The SFTSV RNA transcription, protein synthesis, and progeny virions were reduced in a dose dependent manner by the presence of 6-TG in the in vitro infection assay. Further study on the mechanism of the anti-SFTSV activity showed that 6-TG downregulated the production of early growth response gene-1 (EGR1). Using gene silencing and overexpression, we further confirmed that EGR1 was a host restriction factor against SFTSV. Meanwhile, treatment of infected experimental animals with 6-TG inhibited SFTSV infection and alleviated multi-organ dysfunction. In conclusion, we have identified 6-TG as an effective inhibitor of SFTSV replication via the inhibition of EGR1 expression. Further studies are needed to evaluate of 6-TG as a potential therapeutic for treating SFTS.


Subject(s)
Antiviral Agents , Early Growth Response Protein 1 , Human Umbilical Vein Endothelial Cells , Phlebovirus , Thioguanine , Virus Replication , Animals , Phlebovirus/drug effects , Humans , Virus Replication/drug effects , Thioguanine/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Mice , Vero Cells , Antiviral Agents/pharmacology , Chlorocebus aethiops , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Severe Fever with Thrombocytopenia Syndrome/drug therapy , Severe Fever with Thrombocytopenia Syndrome/virology , Cell Line
15.
Vet Microbiol ; 294: 110106, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38776767

ABSTRACT

Glaesserella parasuis (G. parasuis) is the causative agent of porcine Glässer's disease, resulting in high mortality rates in pigs due to excessive inflammation-induced tissue damage. Previous studies investigating the protective effects of G. parasuis vaccination indicated a possible role of ApoA1 in reflecting disease progression following G. parasuis infection. However, the mechanisms of ApoA1 expression and its role in these infections are not well understood. In this investigation, newborn porcine tracheal (NPTr) epithelial cells infected with G. parasuis were used to elucidate the molecular mechanism and role of ApoA1. The study revealed that the AMPK pathway activation inhibited ApoA1 expression in NPTr cells infected with G. parasuis for the first time. Furthermore, Egr1 was identified as a core transcription factor regulating ApoA1 expression using a CRISPR/Cas9-based system. Importantly, it was discovered that APOA1 protein significantly reduced apoptosis, pyroptosis, necroptosis, and inflammatory factors induced by G. parasuis in vivo. These findings not only enhance our understanding of ApoA1 in response to bacterial infections but also highlight its potential in mitigating tissue damage caused by G. parasuis infection.


Subject(s)
AMP-Activated Protein Kinases , Apolipoprotein A-I , Early Growth Response Protein 1 , Haemophilus parasuis , Signal Transduction , Swine Diseases , Animals , Swine , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Haemophilus parasuis/genetics , Swine Diseases/microbiology , Swine Diseases/genetics , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology , Epithelial Cells/microbiology , Gene Expression Regulation , Trachea/microbiology , Trachea/metabolism , Apoptosis , Animals, Newborn
16.
Environ Int ; 188: 108750, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38788414

ABSTRACT

Extracellular vesicles (EVs) mediate the intercellular crosstalk by transferring functional cargoes. Recently, we have discovered that BaP/BPDE exposure suppresses trophoblast cell migration/invasion and induces miscarriage, which are also regulate by lncRNAs at intracelluar levels. However, the EVs-mediated intercellular regulatory mechanisms are completely unexplored. Specifically, whether EVs might transfer BPDE-induced toxic lncRNA to fresh recipient trophoblast cells and suppress their migration/invasion to further induce miscarriage is completely unknown. In this study, we find that BPDE exposure up-regulates a novel lnc-HZ11, which suppresses EGR1/NF-κB/CXCL12 pathway and migration/invasion of trophoblast cells. Intercellular studies show that EV-HZ11 (lnc-HZ11 in EVs), which is highly expressed in BPDE-exposed donor cells, suppresses EGR1/NF-κB/CXCL12 pathway and migration/invasion in recipient cells by transferring lnc-HZ11 through EVs. Analysis of villous tissues collected from UM (unexplained miscarriage) patients and HC (healthy control) group shows that the levels of BPDE-DNA adducts, lnc-HZ11 or EV-lnc-HZ11, and EGR1/NF-κB/CXCL12 pathway are all associated with miscarriage. Mouse assays show that BaP exposure up-regulates the levels of lnc-Hz11 or EV-Hz11, suppresses Egr1/Nf-κb/Cxcl12 pathway, and eventually induces miscarriage. Knockdown of lnc-Hz11 by injecting EV-AS-Hz11 could effectively alleviate miscarriage in BaP-exposed mice. Furthermore, EV-HZ11 in serum samples could well predict the risk of miscarriage. Collectively, this study not only discovers EVs-HZ11-mediated intercellular mechanisms that BaP/BPDE suppresses trophoblast cell migration/invasion and induces miscarriage but also provides new approach for treatment against unexplained miscarriage through EV-HZ11.


Subject(s)
Abortion, Spontaneous , Cell Movement , Extracellular Vesicles , RNA, Long Noncoding , Trophoblasts , Up-Regulation , Extracellular Vesicles/metabolism , Trophoblasts/metabolism , Humans , Female , RNA, Long Noncoding/genetics , Mice , Animals , Pregnancy , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , NF-kappa B/metabolism
17.
Cancer Lett ; 595: 217000, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-38821254

ABSTRACT

Radiotherapy is one of the predominant treatment modalities for almost all kinds of malignant cancers, including non-small cell lung cancer (NSCLC). Increasing evidence shows that ionizing radiation (IR) induces reactive oxygen species (ROS) leading to lipid peroxidation and subsequently ferroptosis of cancer cells. However, cancer cells evolve multiple mechanisms against ROS biology resulting in resistance to ferroptosis and radiotherapy, of which NRF2 signaling is one of the most studied. In the current research, we identified that microRNA-139 (miR-139) could be a novel radiosensitizer for NSCLC by inhibiting NRF2 signaling. We found that miR-139 possessed great potential as a diagnostic biomarker for NSCLC and multiple other types of cancer. Overexpression of miR-139 increased radiosensitivity of NSCLC cells in vitro and in vivo. MiR-139 directly targeted cJUN and KPNA2 to impair NRF2 signaling resulting in enhanced IR-induced lipid peroxidation and cellular ferroptosis. We proved KPNA2 to be a binding partner of NRF2 that involved in nuclear translocation of NRF2. Moreover, we found that IR induced miR-139 expression through transcriptional factor EGR1. EGR1 bound to the promoter region and transactivated miR-139. Overall, our findings elucidated the effect of EGR1/miR-139/NRF2 in IR-induced ferroptosis of NSCLC cells and provided theoretical support for the potential diagnostic biomarkers and therapeutic targets for the disease.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Early Growth Response Protein 1 , Ferroptosis , Gene Expression Regulation, Neoplastic , Lung Neoplasms , MicroRNAs , NF-E2-Related Factor 2 , Radiation Tolerance , Signal Transduction , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Ferroptosis/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Lung Neoplasms/metabolism , Radiation Tolerance/genetics , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Animals , Cell Line, Tumor , Mice , Male , Reactive Oxygen Species/metabolism , A549 Cells , Mice, Nude , Female
18.
Am J Physiol Cell Physiol ; 326(6): C1648-C1658, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38682237

ABSTRACT

The authors' previous research has shown the pivotal roles of cyclin-dependent kinase 5 (CDK5) and its regulatory protein p35 in nerve growth factor (NGF)-induced differentiation of sympathetic neurons in PC12 cells. During the process of differentiation, neurons are susceptible to environmental influences, including the effects of drugs. Metformin is commonly used in the treatment of diabetes and its associated symptoms, particularly in diabetic neuropathy, which is characterized by dysregulation of the sympathetic neurons. However, the impacts of metformin on sympathetic neuronal differentiation remain unknown. In this study, we investigated the impact of metformin on NGF-induced sympathetic neuronal differentiation using rat pheochromocytoma PC12 cells as a model. We examined the regulation of TrkA-p35/CDK5 signaling in NGF-induced PC12 differentiation. Our results demonstrate that metformin reduces NGF-induced PC12 differentiation by inactivating the TrkA receptor, subsequently inhibiting ERK and EGR1. Inhibition of this cascade ultimately leads to the downregulation of p35/CDK5 in PC12 cells. Furthermore, metformin inhibits the activation of the presynaptic protein Synapsin-I, a substrate of CDK5, in PC12 differentiation. In addition, metformin alters axonal and synaptic bouton formation by inhibiting p35 at both the axons and axon terminals in fully differentiated PC12 cells. In summary, our study elucidates that metformin inhibits sympathetic neuronal differentiation in PC12 cells by disrupting TrkA/ERK/EGR1 and p35/CDK5 signaling. This research contributes to uncovering a novel signaling mechanism in drug response during sympathetic neuronal differentiation, enhancing our understanding of the intricate molecular processes governing this critical aspect of neurodevelopment.NEW & NOTEWORTHY This study unveils a novel mechanism influenced by metformin during sympathetic neuronal differentiation. By elucidating its inhibitory effects from the nerve growth factor (NGF) receptor, TrkA, to the p35/CDK5 signaling pathways, we advance our understanding of metformin's mechanisms of action and emphasize its potential significance in the context of drug responses during sympathetic neuronal differentiation.


Subject(s)
Cell Differentiation , Cyclin-Dependent Kinase 5 , Metformin , Nerve Growth Factor , Neurons , Receptor, trkA , Animals , Metformin/pharmacology , Rats , PC12 Cells , Cyclin-Dependent Kinase 5/metabolism , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Receptor, trkA/metabolism , Receptor, trkA/antagonists & inhibitors , Neurons/drug effects , Neurons/metabolism , Cell Differentiation/drug effects , Signal Transduction/drug effects , Neurogenesis/drug effects , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Phosphotransferases
19.
Cell Mol Biol (Noisy-le-grand) ; 70(3): 125-129, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38650141

ABSTRACT

Myocardial ischemia/reperfusion injury (MIRI) is an irreversible adverse event during the management of coronary heart disease that lacks effective controls. The underlying mechanism of MIRI still requires further investigation. Recent studies have suggested that overexpression of ATF3 protects against MIRI by regulating inflammatory responses, ferroptosis, and autophagy. The downstream target of ATF3, EGR1, also showed cardioprotective properties against MIRI by promoting autophagy. Therefore, further investigating the effect of ATF3/EGR1 pathway on MIRI-induced inflammation and autophagy is needed. Cardiomyocyte MIRI model was established by challenging H9C2 cells with hypoxia/reoxygenation (H/R). The ATF3 overexpression-H/R cell model by transfecting ATF3 plasmid into the H9C2 cell line. The transcription levels of ATF3 and EGR1 were determined using RT-qPCR, the levels of TNF-α and IL-6 were determined using ELISA kits, the protein expression of LC3 I, LC3 II, and P62 was determined via WB, and microstructure of H9C2 cell was observed by transmission electron microscopy (TEM). Overexpression of ATF3 significantly downregulated Egr1 levels, indicating that EGR1 might be the target of ATF3. By upregulating ATF3 levels, the extracellular levels of the inflammatory cytokines TNF-α and IL-6 significantly decreased, and the protein expression of the autophagy markers LC3 I, LC3 II, and P62 significantly increased. TEM results revealed that the cell line in the H/R-ATF3 group exhibited a higher abundance of autophagosome enclosures of mitochondria. The results indicated that ATF3/EGR1 may alleviate inflammation and improve autophagy in an H/R-induced MIRI model of cardiomyocytes.


Subject(s)
Activating Transcription Factor 3 , Autophagy , Early Growth Response Protein 1 , Inflammation , Myocardial Reperfusion Injury , Myocytes, Cardiac , Tumor Necrosis Factor-alpha , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 3/genetics , Autophagy/genetics , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Animals , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Rats , Cell Line , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Interleukin-6/metabolism , Interleukin-6/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Signal Transduction , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics
20.
Biomater Adv ; 160: 213836, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599042

ABSTRACT

The behavior of stem cells is regulated by mechanical cues in their niche that continuously vary due to extracellular matrix (ECM) remodeling, pulsated mechanical stress exerted by blood flow, and/or cell migration. However, it is still unclear how dynamics of mechanical cues influence stem cell lineage commitment, especially in a 3D microenvironment where mechanosensing differs from that in a 2D microenvironment. In the present study, we investigated how temporally varying mechanical signaling regulates expression of the early growth response 1 gene (Egr1), which we recently discovered to be a 3D matrix-specific mediator of mechanosensitive neural stem cell (NSC) lineage commitment. Specifically, we temporally controlled the activity of Ras homolog family member A (RhoA), which is known to have a central role in mechanotransduction, using our previously developed Arabidopsis thaliana cryptochrome-2-based optoactivation system. Interestingly, pulsed RhoA activation induced Egr1 upregulation in stiff 3D gels only, whereas static light stimulation induced an increase in Egr1 expression across a wide range of 3D gel stiffnesses. Actin assembly inhibition limited Egr1 upregulation upon RhoA activation, implying that RhoA signaling requires an actin-involved process to upregulate Egr1. Consistently, static-light RhoA activation rather than pulsed-light activation restricted neurogenesis in soft gels. Our findings indicate that the dynamics of RhoA activation influence Egr1-mediated stem cell fate within 3D matrices in a matrix stiffness-dependent manner.


Subject(s)
Mechanotransduction, Cellular , Neural Stem Cells , rhoA GTP-Binding Protein , rhoA GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/radiation effects , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Light , Cell Differentiation , Humans , Extracellular Matrix/metabolism , Animals
SELECTION OF CITATIONS
SEARCH DETAIL