Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Int J Mol Sci ; 23(9)2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35563251

ABSTRACT

Atopic dermatitis (AD) is one of the most common chronic inflammatory skin diseases worldwide, characterized by intense pruritus and eczematous lesions. Aberrant expression of thymic stromal lymphopoietin (TSLP) in keratinocytes is associated with the pathogenesis of AD and is considered a therapeutic target for the treatment of this disease. Saikosaponin A (SSA) and saikosaponin C (SSC), identified from Radix Bupleuri, exert anti-inflammatory effects. However, the topical effects of SSA and SSC on chronic inflammatory skin diseases are unclear. In this study, we investigated the effects of SSA and SSC on TSLP suppression in an AD-like inflammatory environment. We observed that SSA and SSC suppressed tumor necrosis factor-α-induced TSLP expression by downregulating the expression of the transcription factor early growth response 1 (EGR1) via inhibition of the extracellular signal-regulated kinase 1/2, c-Jun N-terminal kinase 1/2, and p38 mitogen-activated protein kinase pathways. We also confirmed that topical application of SSA or SSC reduced AD-like skin lesions in BALB/c mice challenged with 2,4-dinitrochlorobenzene. Our findings suggest that suppression of EGR1-regulated TSLP expression in keratinocytes might be attributable to the anti-inflammatory effects of SSA and SSC in AD-like skin lesions.


Subject(s)
Dermatitis, Atopic , Skin Diseases , Animals , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/metabolism , Early Growth Response Protein 1/antagonists & inhibitors , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , HaCaT Cells , Humans , Keratinocytes/metabolism , Mice , Oleanolic Acid/analogs & derivatives , Saponins , Skin Diseases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Thymic Stromal Lymphopoietin
2.
Oxid Med Cell Longev ; 2022: 1927260, 2022.
Article in English | MEDLINE | ID: mdl-35126807

ABSTRACT

Increased neutrophil recruitment represents a hallmark event in myocardial ischemia/reperfusion (I/R) injury due to the ensuing inflammatory response. Circular RNAs (circRNAs) are important regulatory molecules involved in cell physiology and pathology. Herein, we analyzed the role of a novel circRNA circ_SMG6 in the regulation of neutrophil recruitment following I/R injury, which may associate with the miR-138-5p/EGR1/TLR4/TRIF axis. Myocardial I/R injury was modeled in vivo by ligation of the left anterior descending (LAD) artery followed by reperfusion in mice and in vitro by exposing a cardiomyocyte cell line (HL-1) to hypoxia/reoxygenation (H/R). Gain- and loss-of-function experiments were performed to evaluate the effect of the circ_SMG6/miR-138-5p/EGR1/TLR4/TRIF axis on cardiac functions, myocardial infarction, myocardial enzyme levels, cardiomyocyte activities, and neutrophil recruitment. We found that the EGR1 expression was increased in myocardial tissues of I/R mice. Knockdown of EGR1 was found to attenuate I/R-induced cardiac dysfunction and infarction area, pathological damage, and cardiomyocyte apoptosis. Mechanistic investigations showed that circ_SMG6 competitively bound to miR-138-5p and consequently led to upregulation of EGR1, thus facilitating myocardial I/R injury in mice and H/R-induced cell injury. Additionally, ectopic EGR1 expression augmented neutrophil recruitment and exacerbated the ensuing I/R injury, which was related to the activated TLR4/TRIF signaling pathway. Overall, our findings suggest that circ_SMG6 may deteriorate myocardial I/R injury by promoting neutrophil recruitment via the miR-138-5p/EGR1/TLR4/TRIF signaling. This pathway may represent a potential therapeutic target in the management of myocardial I/R injury.


Subject(s)
Early Growth Response Protein 1/metabolism , MicroRNAs/metabolism , RNA, Circular/metabolism , Signal Transduction , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Antagomirs/metabolism , Apoptosis , Cell Line , Disease Models, Animal , Early Growth Response Protein 1/antagonists & inhibitors , Early Growth Response Protein 1/genetics , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/veterinary , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neutrophils/immunology , Neutrophils/metabolism , RNA Interference , RNA, Circular/antagonists & inhibitors , RNA, Circular/genetics , RNA, Small Interfering/metabolism , Toll-Like Receptor 4/metabolism
3.
Eur J Pharmacol ; 910: 174470, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34478691

ABSTRACT

Myocardial fibrosis in post-myocardial infarction is a self-healing process of the myocardium, making ventricular remodelling difficult to reverse and develop continuously. Fibroblast growth factor 21 (FGF21) plays an essential role in cardiovascular and metabolic diseases. However, the effect and mechanism of FGF21 action on cardiac inflammation and fibrosis caused by myocardial injury have rarely been reported. Adult male Sprague-Dawley rats administered with or without recombinant human basic FGF21 (rhbFGF21) were assessed using echocardiography and haematoxylin-eosin and Masson's trichrome staining to determine the cardiac function and cardiac inflammation and fibrosis levels. FGF21 might improve cardiac remodelling, as characterised by a decrease in the expression of a series of inflammatory and fibrosis-related factors. Moreover, when FGF receptors (FGFRs) were blocked, the effects of FGF21 disappeared. Mechanistically, we found that oxidative stress induced the downregulation of early growth response protein 1 (EGR1), which contributed to inflammatory factors and fibrosis reduction in cardiomyocytes treated with H2O2. Collectively, FGF21 effectively suppressed the inflammation and fibrosis in post-infarcted hearts by regulating FGFR-EGR1.


Subject(s)
Early Growth Response Protein 1/antagonists & inhibitors , Fibroblast Growth Factors/pharmacology , Myocardial Infarction/drug therapy , Myocardium/pathology , Animals , Cells, Cultured , Disease Models, Animal , Early Growth Response Protein 1/metabolism , Fibroblast Growth Factors/therapeutic use , Fibrosis , Heart/drug effects , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Male , Myocardial Infarction/complications , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardium/immunology , Myocytes, Cardiac , Primary Cell Culture , Rats , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Ventricular Remodeling/drug effects , Ventricular Remodeling/immunology
4.
Neurotox Res ; 39(6): 1981-1990, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34542838

ABSTRACT

Homocysteine (Hcy) is a non-essential amino acid produced from methionine. It has been reported that high concentrations of Hcy are related to the pathogenesis of neurodegenerative diseases and induce the disruption of the blood-brain barrier (BBB) by triggering oxidative stress and inflammation. LCZ696 is a novel antihypertensive agent that has been recently reported to possess promising anti-inflammatory properties. However, whether it has a protective effect on the BBB disruption is still unknown. For the first time, in this study, we aim to investigate whether LCZ696 exerts anti-inflammatory effects on Hcy-induced injury in brain endothelial cells and explore its neuroprotective properties. In in vivo experiments, we found that treatment with LCZ696 ameliorated oxidative stress by reducing malondialdehyde (MDA) and increasing glutathione (GSH). Furthermore, LCZ696 downregulated the excessive release of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) at mRNA and protein levels. Importantly, it reversed the disruption of the BBB induced by Hcy stimulation. In the in vitro human brain microvascular endothelial cell (HBMVEC) experiments, compared to the control, the permeability of the endothelial monolayer was significantly enlarged, the expression level of occludin declined, and Egr-1 upregulated by the introduction of Hcy, and these were all reversed by the treatment with LCZ696. Lastly, we found that the protective effects of LCZ696 against Hcy-induced reduction of occludin and hyper-permeability of the endothelial monolayer were greatly abolished by the overexpression of Egr-1. Taken together, we found that LCZ696 protected against Hcy-induced impairment of BBB integrity by increasing the expression of occludin, all mediated by the inhibition of Egr-1.


Subject(s)
Aminobutyrates/pharmacology , Biphenyl Compounds/pharmacology , Blood-Brain Barrier/drug effects , Early Growth Response Protein 1/antagonists & inhibitors , Homocysteine/adverse effects , Neuroprotective Agents/pharmacology , Occludin/metabolism , Valsartan/pharmacology , Animals , Blotting, Western , Drug Combinations , Early Growth Response Protein 1/metabolism , Homocysteine/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Neuroinflammatory Diseases/chemically induced , Oxidative Stress/drug effects
5.
Mol Neurobiol ; 58(10): 5052-5066, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34245441

ABSTRACT

Postoperative cognitive dysfunction (POCD; cognitive change associated with anesthesia and surgery) is one of the most serious long-term postoperative complications that occur in elderly patients. Dexmedetomidine (DEX) has been shown to be beneficial for improving outcomes of postoperative cognitive function. However, the exact mechanism underlying this role requires is yet to be found. The present study aims to determine the pathways involved in the protective effects of DEX against POCD in C57BL/6 J aged mice. DEX was administered after POCD modeling in C57BL/6 J aged mice. The cognitive function was evaluated after DEX treatment using novel object recognition, open field, and Y-maze tests. We also assessed its effects on neuron apoptosis and production of TNF-α and IL-1ß in mouse brain tissues as well as expression levels of DNA damage-related proteins p53, p21, and γH2AX. Interactions between early growth response 1 (EGR1) and p53, microRNA (miR)-381, and EGR1 were identified by ChIP and luciferase reporter assays, and gain- and loss-of-function experiments were performed to confirm the involvement of their interaction in POCD. DEX administration attenuated hippocampal neuron apoptosis, neuroinflammation, DNA damage, and cognitive impairment in aged mice. miR-381 targeted EGR1 and disrupted its interaction with p53, leading to a decline in hippocampal neuron apoptosis, DNA damage, neuroinflammation, and cognitive impairment. Furthermore, DEX administration resulted in the enhancement of miR-381 expression and the subsequent inhibition of EGR1/p53 to protect against cognitive impairment in aged mice. Overall, these results indicate that DEX may have a potential neuroprotective effect against POCD via the miR-381/EGR1/p53 signaling, shedding light on the mechanisms involved in neuroprotection in POCD.


Subject(s)
Dexmedetomidine/therapeutic use , Early Growth Response Protein 1/metabolism , Hippocampus/metabolism , MicroRNAs/metabolism , Postoperative Cognitive Complications/drug therapy , Postoperative Cognitive Complications/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Dexmedetomidine/pharmacology , Early Growth Response Protein 1/antagonists & inhibitors , Hippocampus/drug effects , Male , Mice , Mice, Inbred C57BL , Postoperative Cognitive Complications/psychology , Tumor Suppressor Protein p53/antagonists & inhibitors
6.
Int J Mol Sci ; 22(9)2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33919431

ABSTRACT

Thymic stromal lymphopoietin (TSLP) is an epithelial cell-derived cytokine that acts as a critical mediator in the pathogenesis of atopic dermatitis (AD). Various therapeutic agents that prevent TSLP function can efficiently relieve the clinical symptoms of AD. However, the downregulation of TSLP expression by therapeutic agents remains poorly understood. In this study, we investigated the mode of action of chrysin in TSLP suppression in an AD-like inflammatory environment. We observed that the transcription factor early growth response (EGR1) contributed to the tumor necrosis factor alpha (TNFα)-induced transcription of TSLP. Chrysin attenuated TNFα-induced TSLP expression by downregulating EGR1 expression in HaCaT keratinocytes. We also showed that the oral administration of chrysin improved AD-like skin lesions in the ear and neck of BALB/c mice challenged with 2,4-dinitrochlorobenzene. We also showed that chrysin suppressed the expression of EGR1 and TSLP by inhibiting the extracellular signal-regulated kinase (ERK) 1/2 and c-Jun N-terminal kinase (JNK) 1/2 mitogen-activated protein kinase pathways. Collectively, the findings of this study suggest that chrysin improves AD-like skin lesions, at least in part, through the downregulation of the ERK1/2 or JNK1/2-EGR1-TSLP signaling axis in keratinocytes.


Subject(s)
Cytokines/metabolism , Early Growth Response Protein 1/antagonists & inhibitors , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Keratinocytes/drug effects , Skin Diseases/drug therapy , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Cytokines/genetics , Dinitrochlorobenzene/toxicity , Humans , Keratinocytes/metabolism , Male , Mice , Mice, Inbred BALB C , Skin Diseases/chemically induced , Skin Diseases/metabolism , Skin Diseases/pathology , Thymic Stromal Lymphopoietin
8.
Mol Biol Rep ; 48(1): 1-11, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33449301

ABSTRACT

Matrix metalloproteinase 1 (MMP-1) initiates the breakdown of matrix networks by cleaving fibrillar collagen during the pathophysiological progression of skin aging. Ageratum houstonianum ethanol extract (AHE) has been used as a traditional herbal medicine to treat external wounds and skin diseases. However, the mechanism of action underlying A. houstonianum-mediated modulation of skin aging has not been investigated. In this study, we evaluated the effect of AHE on MMP-1 expression in HaCaT keratinocytes. Gene expression was analyzed by Reverse transcription-PCR (RT-PCR), Quantitative real-time PCR (Q-PCR), gene promoter-reporter assay, and immunoblotting. We found that AHE abrogated TNFα-induced MMP1 expression at the transcriptional level via the suppression of ERK1/2 mitogen-activated protein kinase (MAPK)-mediated Early Growth Response 1 (EGR1) expression. We also demonstrated that ß-caryophyllene, a cannabinoid receptor 2 (CB2) agonist, is a functional component of the AHE that inhibits TNFα-induced EGR-1 and MMP1 expression. AHE exerts inhibitory activity on TNFα-induced MMP1 expression at the transcription level through EGR-1 downregulation in keratinocytes. ß-Caryophyllene is a bioactive ingredient of AHE that is responsible for the inhibition of TNFα-induced EGR1 expression. ß-Caryophyllene can be used as a potential agent to prevent inflammation-induced skin aging.


Subject(s)
Ageratum/chemistry , Early Growth Response Protein 1/genetics , Matrix Metalloproteinase 1/genetics , Plant Extracts/pharmacology , Skin Aging/drug effects , Early Growth Response Protein 1/antagonists & inhibitors , Gene Expression Regulation/drug effects , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , MAP Kinase Signaling System/drug effects , Plant Extracts/chemistry , Polycyclic Sesquiterpenes/pharmacology , Skin Aging/pathology , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/genetics
9.
Biochem Biophys Res Commun ; 534: 303-309, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33276948

ABSTRACT

Kallikrein-related peptidase 7 (KLK7) is a chymotrypsin-like serine peptidase that plays a crucial role in regulating skin desquamation. KLK7 expression is highly upregulated in atopic dermatitis (AD) skin lesions in both humans and mice. Th2-lymphocyte-derived cytokines, including interleukin (IL)-4 and IL-13, have been shown to promote KLK7 expression in keratinocytes in patients with AD. However, the molecular mechanism underlying KLK7 expression remains poorly understood. Here, we demonstrated that the EGR-1-binding sequence (EBS) in the promoter region of KLK7 played a crucial role in IL-13-induced KLK7 transcription. Disruption of the EBS induced by a point mutation inhibited IL-13-induced KLK7 promoter activity. EGR-1 was shown to directly bind to the EBS, and EGR1 knockdown with shRNA abrogated IL-13-induced KLK7 expression. Using Egr1 knockout mice, we showed that Egr-1 was necessary for KLK7 expression in AD-like lesions induced by the repeated topical application of 2,4-dinitrobenzene on the dorsal skin of mice. We also demonstrated that the ERK1/2 mitogen-activated protein kinase (MAPK) pathway was responsible for EGR-1-dependent KLK7 transcription in response to IL-13 stimulation. Our findings delineate a signaling pathway that contributes to the regulation of KLK7 expression through the IL13-ERK MAPK-EGR1 signaling axis.


Subject(s)
Early Growth Response Protein 1/metabolism , Interleukin-13/metabolism , Kallikreins/genetics , Animals , Dermatitis, Atopic/genetics , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/pathology , Disease Models, Animal , Early Growth Response Protein 1/antagonists & inhibitors , Early Growth Response Protein 1/deficiency , Early Growth Response Protein 1/genetics , Gene Knockdown Techniques , HaCaT Cells , Humans , Kallikreins/metabolism , Keratinocytes/metabolism , Keratinocytes/pathology , MAP Kinase Signaling System , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Trans-Activators/metabolism
10.
BMC Mol Cell Biol ; 21(1): 80, 2020 Nov 10.
Article in English | MEDLINE | ID: mdl-33167868

ABSTRACT

BACKGROUND: Arecoline is an alkaloid natural product found in the areca nut that can induce oral submucous fibrosis and subsequent development of cancer. However, numerous studies have shown that arecoline may inhibit fibroblast proliferation and prevent collagen synthesis. RESULTS: High doses of arecoline (> 32 µg/ml) could inhibit human oral fibroblast proliferation, while low doses of arecoline (< 16 µg/ml) could promote the proliferation of human oral fibroblasts. Wnt5a was found to be both sufficient and necessary for the promotion of fibroblast proliferation. Egr-1 could mediate the expression of Wnt5a in fibroblasts, while NF-κB, FOXO1, Smad2, and Smad3 did not. Treatment with siRNAs specific to Egr-1, Egr inhibitors, or Wnt5a antibody treatment could all inhibit arecoline-induced Wnt5a upregulation and fibroblast proliferation. CONCLUSIONS: Egr-1 mediates the effect of low dose arecoline treatment on human oral mucosa fibroblast proliferation by transactivating the expression of Wnt5a. Therefore, Egr inhibitors and Wnt5a antibodies are potential therapies for treatment of oral submucosal fibrosis and oral cancer.


Subject(s)
Arecoline/adverse effects , Early Growth Response Protein 1/metabolism , Fibroblasts/drug effects , Mouth Mucosa/drug effects , Oral Submucous Fibrosis/metabolism , Wnt-5a Protein/metabolism , Arecoline/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Early Growth Response Protein 1/antagonists & inhibitors , Early Growth Response Protein 1/genetics , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Promoter Regions, Genetic , RNA, Small Interfering , Smad2 Protein/genetics , Smad2 Protein/metabolism , Up-Regulation , Wnt-5a Protein/genetics
11.
Mol Cancer Ther ; 19(11): 2371-2381, 2020 11.
Article in English | MEDLINE | ID: mdl-32847969

ABSTRACT

Direct cell death induction, in addition to immune-effector cell-mediated mechanisms, is one of the key mechanisms of action of anti-CD20 antibodies, and yet the signaling pathways implicated remain poorly investigated. Here we show that the transcription factor EGR-1 is rapidly induced by anti-CD20 antibodies and is a key mediator for CD20-induced cell death. EGR-1 induction results from an increased calcium influx induced by anti-CD20 antibodies. We show that both rituximab and obinutuzumab induce calcium influx, albeit through different mechanisms, and this influx is crucial for cell death induction. Inhibition of the calcium flux with calcium channel blockers (CCB) abolished EGR-1 induction and impaired the efficacy of anti-CD20 antibodies in preclinical in vitro and in vivo models. Finally, we investigated the impact of CCBs in patients treated with anti-CD20 antibodies included in the clinical trials GOYA and REMARC, and found that patients simultaneously receiving CCBs and anti-CD20 therapy have a shorter progression-free survival and overall survival. These results reveal EGR-1 as a key mediator of the direct cytotoxic activity of anti-CD20 antibodies and provide a rationale to evaluate EGR-1 expression as a new biomarker to predict response to anti-CD20 treatment. In addition, our findings show that calcium influx is required for anti-CD20-mediated tumor cell death and suggest that simultaneous administration of calcium channel blocking agents could be deleterious in patients receiving anti-CD20-based immunotherapy.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Calcium Channel Blockers/pharmacology , Drug Antagonism , Early Growth Response Protein 1/antagonists & inhibitors , Rituximab/pharmacology , Animals , Antigens, CD20 , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Early Growth Response Protein 1/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/diagnosis , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/mortality , Mice , NIH 3T3 Cells , Prognosis , Signal Transduction , Treatment Outcome , Xenograft Model Antitumor Assays
12.
Aging (Albany NY) ; 12(13): 12684-12702, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32629426

ABSTRACT

Long non-coding RNAs (lncRNAs) have been largely reported to contribute to the development and progression of abdominal aortic aneurysm (AAA), a common vascular degenerative disease. The present study was set out with the aim to investigate the possible role of lncRNA Sox2ot in the development of AAA. In this study, we found that lncRNA Sox2ot and early growth response factor-1 (Egr1) were highly expressed, while microRNA (miR)-145 was poorly expressed in Ang II-induced AAA mice and oxidative stress-provoked vascular smooth muscle cell (VSMC) model. Egr1 was a potential target gene of miR-145, and lncRNA Sox2ot could competitively bind to miR-145 to upregulate Egr1 expression. Overexpression of miR-145-5p was found to attenuate oxidative stress and inflammation by inhibiting Egr1 both in vivo and in vitro, which was counteracted by lncRNA Sox2ot. Taken together, the present study provides evidence that downregulation of lncRNA Sox2ot suppressed the expression of Egr1 through regulating miR-145, thus inhibiting the development of AAA, highlighting a theoretical basis for AAA treatment.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Early Growth Response Protein 1/metabolism , MicroRNAs/metabolism , Oxidative Stress/genetics , RNA, Long Noncoding , Animals , Aorta, Abdominal/cytology , Aorta, Abdominal/metabolism , Apolipoproteins E/genetics , Cells, Cultured , Down-Regulation , Early Growth Response Protein 1/antagonists & inhibitors , Gene Silencing , Inflammation , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Oxidative Stress/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
13.
Mol Med Rep ; 22(3): 2496-2506, 2020 09.
Article in English | MEDLINE | ID: mdl-32705196

ABSTRACT

Acute pancreatitis (AP) is a common digestive disorder with high morbidity and mortality. The present study aimed to investigate the expression of early growth response protein 1 (Egr1), and the effect of competing endogenous (ce)RNA network on trypsinogen activation. Pancreatic acinar intracellular trypsinogen activation (PAITA) is an important event in the early stage of AP; however, the underlying mechanisms remain unclear. The present study used taurolithocholic acid 3­sulfate (TLC­S)­treated AR42J cells (pancreatic cell line) to establish a PAITA model. A gene microarray and bioinformatics analysis was performed to identify the potential key targets in PAITA. The results demonstrated that Egr1, an important transcription factor, was significantly overexpressed in PAITA. In Egr1 small interfering (si)RNA­transfected cells, Egr1 expression was decreased and trypsinogen activation was significantly decreased compared with negative control siRNA­transfected cells, indicating that in TLC­S­induced PAITA, overexpression of Egr1 enhanced trypsinogen activation. A ceRNA network [mRNA­microRNA (miRNA/miR)­long non­coding (lnc)RNA] generated using the PAITA model revealed that the effects of Egr1 on PAITA may be regulated by multiple ceRNA pairs, and the lncRNAs (including NONRATT022624 and NONRATT031002) and miRNAs [including Rattus norvegicus (rno)­miR­214­3p and rno­miR­764­5p] included in the ceRNA pairs may serve roles in PAITA by regulating the expression of Egr1. The results of the present study may provide novel targets for researching the underlying mechanisms of, and developing treatments for AP.


Subject(s)
Early Growth Response Protein 1/genetics , Gene Expression Profiling/methods , Gene Regulatory Networks , Pancreatitis/genetics , Taurolithocholic Acid/analogs & derivatives , Trypsinogen/metabolism , Animals , Cell Line , Computational Biology , Early Growth Response Protein 1/antagonists & inhibitors , Enzyme Activation/drug effects , MicroRNAs/genetics , Models, Biological , Oligonucleotide Array Sequence Analysis , Pancreatitis/chemically induced , Pancreatitis/metabolism , RNA, Long Noncoding/genetics , RNA, Small Interfering/pharmacology , Rats , Taurolithocholic Acid/adverse effects , Up-Regulation/drug effects
14.
Mol Biol Rep ; 47(8): 5953-5962, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32705506

ABSTRACT

Pro-opiomelanocortin (POMC) is a large precursor protein of and ß-endorphin. POMC expressed in keratinocytes regulates various pathophysiological responses, such as pruritus in atopic dermatitis. Interleukin (IL)-31 is a T helper 2 (Th2)-derived cytokine that functions as a pruritogen, stimulating the sensory neurons in the skin. However, the regulatory mechanism underlying IL-31-induced POMC expression in keratinocytes remains largely unknown. Herein, using a 5'-serial deletion and site-specific mutation constructs of the regulatory region of POMC, we demonstrated that a putative EGR1-binding sequence (EBS) motif in POMC is required for its upregulation by IL-31 in HaCaT keratinocytes. Notably, EGR-1 directly interacted with the EBS motif in POMC. The ectopic expression of EGR-1 stimulated the POMC promoter activity, whereas the knockdown of EGR-1 expression by RNA interference reduced IL-31-induced POMC expression. Furthermore, we observed that three major mitogen-activated protein kinases, ERK, JNK, and p38 kinase, mediated IL-31-induced EGR-1 expression. In summary, our results suggest that EGR-1 trans-activates POMC in response to IL-31 stimulation in HaCaT keratinocytes.


Subject(s)
Early Growth Response Protein 1/physiology , Interleukins/pharmacology , Keratinocytes/metabolism , Pro-Opiomelanocortin/genetics , Transcription, Genetic/drug effects , Amino Acid Motifs , Cell Line, Transformed , Early Growth Response Protein 1/antagonists & inhibitors , Early Growth Response Protein 1/genetics , Genes, Reporter , Genes, Synthetic , Humans , MAP Kinase Signaling System/drug effects , Mutagenesis, Site-Directed , Point Mutation , Pro-Opiomelanocortin/biosynthesis , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Transcriptional Activation , Up-Regulation/drug effects
15.
Mol Immunol ; 120: 61-66, 2020 04.
Article in English | MEDLINE | ID: mdl-32078859

ABSTRACT

Myocardial infarction (MI) or heart attack is a deadly event with high prevalence. In the present study, we investigated the effects of the polypeptide copolymer glatiramer acetate (GA) in H9c2 rat cardiomyocytes exposed to oxygen-glucose deprivation/reperfusion injury. Immediately following MI, an acute inflammatory response is triggered that causes activation of various proinflammatory cytokines, infiltration of immune cells, and neovascularization. This response is largely mediated by some genes such as TNF-α, IL-6, ICAM-1, and VEGF. Additionally, the rapid influx of oxidants, such as reactive oxygen species (ROS), leads to a harmful state of oxidative stress. Here, we found that GA could reduce OGD/R-induced inflammation and oxidative stress by inhibiting the expression of TNF-α, IL-6, ICAM-1, and VEGF, and suppressing the production of ROS via reduced NADPH oxidase 1 (NOX1) expression. To elucidate the pathways involved in these promising results, we took a close look at the impact of the endothelial growth response-1 (Egr-1), a transcriptional factor recognized as a mediator of MI-related inflammation and cellular injury. Using siRNA for Egr-1, we found that GA could reduce the expression of ICAM-1 and VEGF by inhibiting Egr-1 expression. Together, our findings indicate a novel therapeutic potential of GA in the treatment of MI.


Subject(s)
Cardiotonic Agents/pharmacology , Early Growth Response Protein 1/antagonists & inhibitors , Glatiramer Acetate/pharmacology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Animals , Cell Line , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Glucose/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/drug therapy , Oxidative Stress/drug effects , Oxygen/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Reactive Oxygen Species/metabolism
16.
Int J Mol Sci ; 21(1)2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31906413

ABSTRACT

Muscle invasive bladder carcinoma is a highly malignant cancer with a high mortality rate, due to its tendency to metastasize. The tyrosine kinase recepteur d'origine nantais (RON) promotes bladder carcinoma metastasis. Lysophosphatidic acid (LPA) is a phospholipid derivative, which acts as a signaling molecule to activate three high affinity G-protein coupled receptors, LPA1, LPA2, and LPA3. This in turn leads to cell proliferation and contributes to oncogenesis. However, little is known about the effects of LPA on invasive bladder cancer (IBC). In this study, we discovered that LPA upregulated RON expression, which in turn promoted cell invasion in bladder cancer T24 cells. As expected, we found that the LPA receptor was essential for the LPA induced increase in RON expression. More interestingly, we discovered that LPA induced RON expression via the MAPK (ERK1/2, JNK1/2), Egr-1, AP-1, and NF-κB signaling axes. These results provide experimental evidence and novel insights regarding bladder malignancy metastasis, which could be helpful for developing new therapeutic strategies for IBC treatment.


Subject(s)
Cell Movement/drug effects , Lysophospholipids/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Urinary Bladder Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Early Growth Response Protein 1/antagonists & inhibitors , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Humans , NF-kappa B/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Urinary Bladder Neoplasms/metabolism
17.
BMC Nephrol ; 20(1): 245, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31277592

ABSTRACT

BACKGROUND: It has been proposed that peroxisome proliferator-activated receptor-γ (PPARγ) agonists might reduce renal fibrosis, however, several studies had contradictory results. Moreover, the possible interaction of TGF-ß1, PPARγ, and transcription factors in renal fibrosis have not been investigated. We hypothesized that oral pioglitazone treatment would inhibit TGF-ß-driven renal fibrosis and its progression, by modulating profibrotic transcription factors in TGF-ß1 transgenic mice. METHODS: Male C57Bl/6 J mice (control, CTL, n = 14) and TGF-ß overexpressing transgenic mice (TGFß, n = 14, having elevated plasma TGF-ß1 level) were divided in two sets at 10 weeks of age. Mice in the first set were fed with regular rodent chow (CTL and TGFß, n = 7/group). Mice in the second set were fed with chow containing pioglitazone (at a dose of 20 mg/kg/day, CTL + Pio and TGFß+Pio, n = 7/group). After 5 weeks of treatment, blood pressure was assessed and urine samples were collected, and the kidneys were analyzed for histology, mRNA and protein expression. RESULTS: TGF-ß1 induced glomerulosclerosis and tubulointerstitial damage were significantly reduced by pioglitazone. Pioglitazone inhibited renal mRNA expression of all the profibrotic effectors: type-III collagen, TGF-ß1, CTGF and TIMP-1, and alike transcription factors cFos/cJun and protein expression of EGR-1, and STAT3 protein phosphorylation. CONCLUSIONS: Oral administration of PPARγ agonist pioglitazone significantly reduces TGF-ß1-driven renal fibrosis, via the attenuation of EGR-1, STAT3 and AP-1. This implies that PPARγ agonists might be effective in the treatment of chronic kidney disease patients.


Subject(s)
Early Growth Response Protein 1/antagonists & inhibitors , Kidney Diseases/prevention & control , PPAR gamma/agonists , Pioglitazone/therapeutic use , STAT3 Transcription Factor/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Early Growth Response Protein 1/metabolism , Fibrosis , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pioglitazone/pharmacology , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta/toxicity
18.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2490-2503, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31201921

ABSTRACT

Osteoarthritis is one of the most common orthopedic diseases in elderly people who have lost their mobility. In this study,we observed abnormally high EGR1 expression in the articular cartilage of patients with osteoarthritis. We also found significantly high EGR1 expression in the articular cartilage of mice with destabilized medial meniscus (DMM)-induced osteoarthritis and 20-month-old mice. In vitro experiments indicated that IL-1ß could significantly enhance EGR1 expression in primary mouse chondrocytes. EGR1 over-expression in chondrocytes using adenovirus could inhibit COl2A1 expression and enhance MMP9 and MMP13 expression. And silencing EGR1, using RNAi, had the opposite effects. Moreover, EGR1 over-expression accelerated chondrocyte hypertrophy in vitro, and EGR1 knockdown reversed this effect. We then explored the underlying mechanism. EGR1 over-expression increased Kruppel-Like Factor 5 (KLF5) protein level without influencing its synthesis. Enhanced EGR1 expression induced its integration with KLF5, leading to suppressed ubiquitination of KLF5. Moreover, EGR1 prompted ß-catenin nuclear transportation to control chondrocyte hypertrophy. Ectopic expression of EGR1 in articular cartilage aggravated the degradation of the cartilage matrix in vivo. The EGR1 inhibitor, ML264, protected chondrocytes from IL-1ß-mediated cartilage matrix degradation in vitro and DMM-induced osteoarthritis in vivo. Above all, we demonstrate the effect and mechanisms of EGR1 on osteoarthritis and provide evidence that the ML264 might be a potential drug for treating osteoarthritis in the future.


Subject(s)
Cartilage, Articular/metabolism , Early Growth Response Protein 1/metabolism , Kruppel-Like Transcription Factors/metabolism , beta Catenin/metabolism , Aging , Animals , Cartilage, Articular/pathology , Chondrocytes/cytology , Chondrocytes/metabolism , Early Growth Response Protein 1/antagonists & inhibitors , Early Growth Response Protein 1/genetics , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Interleukin-1beta/pharmacology , Kruppel-Like Transcription Factors/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Osteoarthritis/pathology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Ubiquitin/metabolism , Ubiquitination , Up-Regulation/drug effects , beta Catenin/antagonists & inhibitors
19.
Talanta ; 202: 591-599, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31171226

ABSTRACT

Photodynamic therapy (PDT) was considered as an effective treatment. Whereas only PDT is not enough to achieve effective therapy on account of irradiation intensity decreases as depth increases as well as tumor hypoxia. Combination with gene therapy and photodynamic therapy have emerged as an effective strategy to improve therapeutic effectiveness. In the present study, a GSH responsive MnO2 was employed to delivery TB and DNAzyme for cancer imaging and PDT-gene combination treatment. TB, a photosensiters with aggregation-induced emission characteristic, was employed for photodynamic therapy, while DNAzyme, acting as catalysts for the degradation of EGR-1 mRNA, was exploited for gene silencing. All of the results of tumor treatment in vitro have implied that MnO2-DNAzyme-TB nanocomposite (MDT) can internalize into cells. Subsequently, MDT could decrease the expression of EGR-1 by gene silencing that enabling inhibition of cell growth. In addition, the singlet oxygen which was generated by the aggregated TB were able to further suppress cell growth. Combination therapy of photodynamic as well as gene therapy greatly enhanced antitumor efficiencies. Furthermore, in vivo tumor treatment experiments demonstrated that MDT under illumination can effectively inhibit the tumor growth of MCF-7 tumor-bearing mice by photodynamic and gene silencing combination therapy.


Subject(s)
Antineoplastic Agents/pharmacology , DNA, Catalytic/metabolism , Genetic Therapy , Manganese Compounds/pharmacology , Oxides/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA, Catalytic/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Early Growth Response Protein 1/antagonists & inhibitors , Early Growth Response Protein 1/genetics , Female , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Manganese Compounds/chemical synthesis , Manganese Compounds/chemistry , Mice , Mice, Nude , Nanocomposites , Optical Imaging , Oxides/chemical synthesis , Oxides/chemistry , Particle Size , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , Structure-Activity Relationship , Surface Properties
20.
Arthritis Res Ther ; 21(1): 92, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30971291

ABSTRACT

BACKGROUND: This study aimed to explore the molecular mechanism and clinical relevance of iguratimod in the regulation of human B cell terminal differentiation. METHODS: An in vitro human antibody-secreting cell (ASC) differentiation system was established to test the effect of iguratimod. B cell phenotype and key transcription factors (TFs) relevant to ASC differentiation were analyzed through flow cytometry and qPCR. The COX-2 activity was measured by enzyme immunoassay (EIA). RNA sequencing was used to identify potential targets of iguratimod. We enrolled six treatment-naive rheumatoid arthritis (RA) patients whose blood samples were collected for phenotypic and molecular studies along with 12-week iguratimod monotherapy. RESULTS: Iguratimod inhibited human ASC generation without affecting B cell activation and proliferation. Iguratimod showed only weak COX-2 activity. Gene set enrichment analysis (GSEA) identified that protein kinase C (PKC) pathway was targeted by iguratimod which was confirmed by PKC activity detection. Furthermore, early growth response 1 (EGR1), a target of PKC and a non-redundant TF for ASC differentiation, was found to be the most downregulated gene in iguratimod-treated B cells. Lastly, iguratimod monotherapy decreased peripheral ASCs and was associated with improved disease activity. The expression of major ASC-related TFs, including EGR1, was similarly downregulated in patient blood samples. CONCLUSIONS: Iguratimod inhibits ASC differentiation both in vitro and in RA patients. Our study suggests that PKC/EGR1 axis, rather than COX-2, is critically involved in the inhibitory effect by iguratimod on human ASC differentiation. Iguratimod could have a broader application to treat B cell-related autoimmune diseases in clinics.


Subject(s)
Antirheumatic Agents/pharmacology , B-Lymphocytes/drug effects , Cell Differentiation/drug effects , Chromones/pharmacology , Early Growth Response Protein 1/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Sulfonamides/pharmacology , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/drug therapy , B-Lymphocytes/metabolism , Cell Differentiation/physiology , Cells, Cultured , Chromones/therapeutic use , Early Growth Response Protein 1/blood , Humans , Protein Kinase C/blood , Sulfonamides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...