Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Hum Mol Genet ; 31(4): 638-650, 2022 02 21.
Article in English | MEDLINE | ID: mdl-34590683

ABSTRACT

Activated neutrophil-derived exosomes reportedly contribute to the proliferation of airway smooth muscle cells (ASMCs), thereby aggravating the airway wall remodeling during asthma; however, the specific mechanism remains unclear. Lipopolysaccharide (LPS)-EXO and si-CRNDE-EXO were extracted from the media of human neutrophils treated with LPS and LPS + si-CRNDE (a siRNA targets long non-coding RNA CRNDE), respectively. Human ASMCs were co-cultured with LPS-EXO or si-CRNDE-EXO, and cell viability, proliferation and migration were measured. The interplay of colorectal neoplasia differentially expressed (CRNDE), inhibitor of nuclear factor kappa B kinase subunit beta (IKKß) and nuclear receptor subfamily 2 group C member 2 (TAK1) was explored using RNA immunoprecipitation (RIP) and Co-IP assays. A mouse model of asthma was induced using ovalbumin. CRNDE was upregulated in LPS-EXO and successfully transferred from LPS-treated neutrophils to ASMCs through exosome. Mechanically, CRNDE loaded in LPS-EXO reinforced TAK1-mediated IKKß phosphorylation, thereby activating the nuclear factor kappa B (NF-κB) pathway. Functionally, silencing CRNDE in LPS-EXO, an IKKß inhibitor, and an NF-κB inhibitor all removed the upregulation of cell viability, proliferation and migration induced by LPS-EXO in ASMCs. In the end, the in vivo experiment demonstrated that CRNDE knockdown in neutrophils effectively reduced the thickness of bronchial smooth muscle in a mouse model for asthma. Activated neutrophils-derived CRNDE was transferred to ASMCs through exosomes and activated the NF-κB pathway by enhancing IKKß phosphorylation. The latter promoted the proliferation and migration of ASMCs and then contributed to airway remodeling in asthma.


Subject(s)
Asthma , Colorectal Neoplasms , RNA, Long Noncoding , Airway Remodeling , Animals , Asthma/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Disease Models, Animal , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Lipopolysaccharides/pharmacology , Mice , Myocytes, Smooth Muscle/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neutrophils/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
2.
Skin Res Technol ; 30(7): e13842, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38965799

ABSTRACT

BACKGROUND: As the most important modifications on the RNA level, N6-methyladenosine (m6A-) and 5-methylcytosine (m5C-) modification could have a direct influence on the RNAs. Long non-coding RNAs (lncRNAs) could also be modified by methylcytosine modification. Compared with mRNAs, the function of lncRNAs could be more potent to some extent in biological processes like tumorigenesis. Until now, rare reports have been done associated with cutaneous melanoma. Herein, we wonder if the m6A- and m5C- modified lncRNAs could influence the immune landscape and prognosis in melanoma, and we also want to find some lncRNAs which could directly affect the malignant behaviors of melanoma. METHODS: Systematically, we explored the expression pattern of m6A- and m5C- modified lncRNAs in melanoma from datasets including UCSC Xena and NCBI GEO, and the prognostic lncRNAs were selected. Then, according to the expression pattern of lncRNAs, melanoma samples from these datasets were divided into several subtypes. Prognostic model, nomogram survival model, drug sensitivity, GO, and KEGG pathway analysis were performed. Furthermore, among several selected lncRNAs, we identified one lncRNA named LINC00893 and investigated its expression pattern and its biological function in melanoma cell lines. RESULTS: We identified 27 m6A- and m5C- related lncRNAs which were significantly associated with survival, and we made a subtype analysis of melanoma samples based on these 27 lncRNAs. Among the two subtypes, we found differences of immune cells infiltration between these two subtypes. Then, LASSO algorithm was used to screen the optimized lncRNAs combination including ZNF252P-AS1, MIAT, FAM13A-AS1, LINC-PINT, LINC00893, AGAP2-AS1, OIP5-AS1, and SEMA6A-AS1. We also found that there was a significant correlation between the different risk groups predicted based on RS model and the actual prognosis. The nomogram survival model based on independent survival prognostic factors was also constructed. Besides, sensitivity to chemotherapeutic agents, GO and KEGG analysis were performed. In different risk groups, a total of 14 drug molecules with different distributions were obtained, which included AZD6482, AZD7762, AZD8055, camptothecin, dasatinib, erlotinib, gefitinib, gemcitabine, GSK269962A, nilotinib, rapamycin, and sorafenib. A total of 55 significantly related biological processes and 17 KEGG signaling pathways were screened. At last, we noticed that LINC00893 had a relatively lower expression in melanoma tissue and cell lines compared with adjacent tissues and epidermal melanocyte, and down-regulation of LINC00893 could promote the malignant behavior of melanoma cells in A875 and MV3. In these two melanoma cell lines, down-regulation of m6A-related molecules like YTHDF3 and METTL3 could promote the expression of LINC00893. CONCLUSION: We made an analysis of m6A- and m5C- related lncRNAs in melanoma samples and a prediction of these lncRNAs' role in prognosis, tumor microenvironment, immune infiltration, and clinicopathological features. We also found that LINC00893, which is potentially regulated by m6A modification, could serve as a tumor-suppressor in melanoma and play an inhibitory role in melanoma metastasis.


Subject(s)
Adenosine , Melanoma , RNA, Long Noncoding , Skin Neoplasms , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Melanoma/genetics , Melanoma/pathology , Melanoma/mortality , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/mortality , Adenosine/analogs & derivatives , Adenosine/metabolism , Prognosis , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Melanoma, Cutaneous Malignant , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Nomograms
3.
BMC Plant Biol ; 22(1): 43, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35062882

ABSTRACT

In desert habitats, sand burial is an important factor affecting germination of plant seeds and seedling growth. Xanthium spinosum has strong adaptability in arid desert areas, and is a common malignant invasive plant in Xinjiang, China. The effects of different sand burial depths on seed germination, seedling emergence, growth and biomass allocation were studied to provide a scientific basis for further control of X. spinosum. Six sand burial depths (1, 2, 3, 5, 7 and 9 cm) were established to explore the response of X. spinosum seed germination and seedling growth to sand burial. The first emergence time, peak emergence time, emergence rate, seedling growth height, biomass and biomass distribution of X. spinosum seeds was significantly different at sand burial depths (P < 0.05). The X. spinosum seeds had the highest emergence rate (71.5%) at 1 cm sand burial and the maximum seedling height (7.1 cm). As sand burial depth increased, the emergence rate and seedling height gradually decreased. Emergence rate (12.25%) and seedling height (2.9 cm) were lowest at 9 cm sand burial. The root length at 9 cm depth (13.6 cm) was significantly higher than that at other sand depths (P < 0.05). The sand burial depth affected the biomass accumulation and distribution of X. spinosum. As sand burial depth increased, the root biomass and rhizome ratio increased, and the most deeply buried seedlings allocated more biomass for root growth. The optimal sand burial depth for seed germination and seedling growth of X. spinosum was 1-3 cm, and high burial depth (5-9 cm) was not conducive to the germination and growth of X. spinosum seedlings. For prevention and control of X. spinosum, we suggest deeply ploughing crops before sowing to ensure X. spinosum seeds are ploughed into a deep soil layer.


Subject(s)
Sand , Seedlings/growth & development , Seeds/growth & development , Xanthium/growth & development , Biomass , China , Germination/physiology , Introduced Species
4.
Pulm Pharmacol Ther ; 77: 102173, 2022 12.
Article in English | MEDLINE | ID: mdl-36280202

ABSTRACT

BACKGROUND: Asthma is an inflammatory syndrome characterized by airway hyperresponsiveness, bronchial inflammation, and airway remodeling. Abnormal proliferation of airway smooth muscle cells (ASMCs) is the main pathological feature of asthma. This study investigated the function and mechanism of serine arginine-rich splicing factor 1 (SRSF1) in ASMC proliferation in asthma. METHODS: SRSF1 expressions in the bronchi of ovalbumin-induced asthmatic mice and IgE-treated mouse ASMCs (mASMCs) were evaluated using quantitative real-time PCR and Western blot. The localization and expression of SRSF1 in the bronchi of asthmatic mice were assessed by immunohistochemistry. Functionally, gain- and loss-of-function assays, flow cytometry, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays were conducted. Mechanistically, RNA degradation assay, RNA immunoprecipitation, RNA pull-down, and dual-luciferase reporter gene assays were carried out. RESULTS: SRSF1 was highly expressed in the bronchi of ovalbumin-induced asthma mice and IgE-treated mASMCs and was mainly located in the nucleus. Experiments on the function of SRSF1 showed that the silencing of SRSF1 induced the cell cycle of mASMC arrest and restrained mASMC proliferation. Investigations into the mechanism of SRSF1 revealed that SRSF1 and miR-135a are competitively bound to the 3'UTR region of Cyclin D2 (CCND2). SRSF1 overexpression repressed the degradation of CCND2 mRNA, and miR-135a negatively regulated CCND2 expression. Furthermore, SRSF1 knockdown inhibited ASMC proliferation in asthma mouse models by regulating the levels of miR-135a and CCND2. CONCLUSION: SRSF1 knockdown repressed ASMC proliferation in asthma by regulating miR-135a/CCND2 levels.


Subject(s)
Asthma , Cyclin D2 , MicroRNAs , Serine-Arginine Splicing Factors , Animals , Mice , Asthma/genetics , Asthma/pathology , Bronchi/metabolism , Cell Proliferation/genetics , Cyclin D2/metabolism , Immunoglobulin E , MicroRNAs/genetics , Myocytes, Smooth Muscle/metabolism , Ovalbumin , Serine-Arginine Splicing Factors/metabolism
5.
Ann Allergy Asthma Immunol ; 129(3): 360-365.e1, 2022 09.
Article in English | MEDLINE | ID: mdl-35598882

ABSTRACT

BACKGROUND: Stevens-Johnson syndrome/toxic epidermal necrolysis (SJS/TEN) is a severe cutaneous adverse reaction to drugs with considerable morbidity and mortality. Immunomodulators for SJS/TEN including systemic corticosteroids and intravenous immunoglobulin (IVIG) have been widely used in clinical practice. Emerging evidence suggested the therapeutic effects of tumor necrosis factor-α antagonists on SJS/TEN. OBJECTIVE: To compare the efficacy and safety of IVIG and systemic steroids in conjunction with or without etanercept, a tumor necrosis factor-α inhibitor, for patients with SJS/TEN. METHODS: We undertook a retrospective review of 41 patients with SJS/TEN admitted to our institution from 2015 to February 2021. A total of 25 patients with integrated data were involved in this study, of which 14 patients were treated with IVIG and corticosteroids and 11 were in addition given etanercept. The clinical characteristics, duration of hospitalization, exposure time to high-dose steroids, and the total amount of systemic steroids were analyzed. RESULTS: In comparison to conventional therapy, conjunction with etanercept reduced the duration of hospitalization (13.5 vs 19.0 days; P = .01), the exposure time of high-dose steroids (7.1 vs 14.9 days; P = .01), and the overall amount of systemic steroid (925 mg vs 1412.5 mg; P = .03) in patients with SJS/TEN. No pronounced adverse effects were observed within 6 months of follow-up after the treatment. CONCLUSION: The add-in of etanercept at the time of initiating conventional therapy could be a superior option to accelerate disease recovery and reduce the high dose and total amount of systemic steroids without pronounced adverse events in patients with SJS/TEN.


Subject(s)
Etanercept , Stevens-Johnson Syndrome , Adrenal Cortex Hormones/therapeutic use , Etanercept/therapeutic use , Humans , Immunoglobulins, Intravenous/therapeutic use , Retrospective Studies , Steroids/therapeutic use , Stevens-Johnson Syndrome/drug therapy , Tumor Necrosis Factor Inhibitors/therapeutic use
6.
J Immunol ; 202(4): 1176-1185, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30642978

ABSTRACT

Low-molecular mass protein 7 (LMP7) is a proteolytic subunit of the immunoproteasome that is involved in regulating inflammatory responses. However, the role of LMP7 in the pathogenesis of abdominal aortic aneurysm (AAA) remains unknown. In this study, ApoE knockout (KO) or LMP7/ApoE double KO (dKO) mice were infused with angiotensin II (Ang II, 1000 ng/kg per minute) for up to 28 d. We found that LMP7 expression was significantly upregulated in AAA tissues from ApoE KO mice and human patients. Moreover, Ang II infusion markedly increased the incidence and severity of AAA in ApoE KO mice, which was considerably reduced in LMP7/ApoE dKO mice. Histological alterations, including aortic wall thickening, collagen deposition, elastin fragmentation, and vascular smooth muscle cell apoptosis in AAA tissue of ApoE KO mice, were also significantly attenuated in LMP7/ApoE dKO mice. Interestingly, LMP7/ApoE dKO mice showed a marked reduction of infiltration of CD3+ T cells, especially CD4+ T cells in AAA tissues compared with ApoE KO mice. Moreover, ablation of LMP7 substantially inhibited the differentiation of CD4+ T cells into Th1 and Th17 cells by reducing the activation of multiple transcriptional factors. We also investigated the effects of an LMP7-specific inhibitor PR-957 (also known as ONX 0914) on AAA formation in ApoE KO mice. PR-957 treatment could reduce the AAA incidence and severity. In conclusion, our results provide, to our knowledge, novel evidence that ablation or pharmacological inhibition of LMP7 attenuates Ang II-induced AAA formation, and LMP7 might be a novel therapeutic target for treating AAA in humans.


Subject(s)
Aortic Aneurysm, Abdominal/drug therapy , Aortic Aneurysm, Abdominal/prevention & control , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Animals , Aortic Aneurysm, Abdominal/metabolism , Biocatalysis , Inflammation/drug therapy , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1 Cells , Th17 Cells
7.
Am J Dermatopathol ; 43(5): 373-376, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33481374

ABSTRACT

ABSTRACT: Chronic arsenism usually occurs after a long-term unawareness of arsenic exposure from environment, occupation, food, and water. We here reported 3 cases with diffused arsenic keratosis and skin cancers derived from long-term arsenic medication ingestion. In these cases, hyperkeratotic skin lesions were initially found on palms and soles, slowly progressed to every part of the skin and lasted maximally for over 30 years. Skin cancers were diagnosed and removed intermittently within decades, but with no malignancies in other organs. Oral retinoids combing with topical 5- fluorouracil and photodynamic treatment yielded a desirable outcome.


Subject(s)
Arsenic Poisoning/pathology , Iatrogenic Disease , Keratoderma, Palmoplantar/chemically induced , Skin Neoplasms/chemically induced , Aged, 80 and over , Humans , Male , Middle Aged
8.
J Cell Mol Med ; 24(11): 6233-6241, 2020 06.
Article in English | MEDLINE | ID: mdl-32343488

ABSTRACT

The pathogenesis of cardiac hypertrophy is tightly associated with activation of intracellular hypertrophic signalling pathways, which leads to the synthesis of various proteins. Tripartite motif 10 (TRIM10) is an E3 ligase with important functions in protein quality control. However, its role in cardiac hypertrophy was unclear. In this study, neonatal rat cardiomyocytes (NRCMs) and TRIM10-knockout mice were subjected to phenylephrine (PE) stimulation or transverse aortic constriction (TAC) to induce cardiac hypertrophy in vitro and in vivo, respectively. Trim10 expression was significantly increased in hypertrophied murine hearts and PE-stimulated NRCMs. Knockdown of TRIM10 in NRCMs alleviated PE-induced changes in the size of cardiomyocytes and hypertrophy gene expression, whereas TRIM10 overexpression aggravated these changes. These results were further verified in TRIM10-knockout mice. Mechanistically, we found that TRIM10 knockout or knockdown decreased AKT phosphorylation. Furthermore, we found that TRIM10 knockout or knockdown increased ubiquitination of phosphatase and tensin homolog (PTEN), which negatively regulated AKT activation. The results of this study reveal the involvement of TRIM10 in pathological cardiac hypertrophy, which may occur by prompting of PTEN ubiquitination and subsequent activation of AKT signalling. Therefore, TRIM10 may be a promising target for treatment of cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tripartite Motif Proteins/metabolism , Animals , Aorta/pathology , Cardiomegaly/pathology , Constriction, Pathologic , Intracellular Signaling Peptides and Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Proteolysis , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Tripartite Motif Proteins/deficiency , Ubiquitination
9.
Biochem Biophys Res Commun ; 533(4): 1012-1020, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33019975

ABSTRACT

Macrophages contribute to abdominal aortic aneurysm (AAA), but the effect of macrophage on AAA formation is not totally understood. Recent research proved that macrophage pyroptosis plays an important role in many cardiovascular disease. However, whether macrophage pyroptosis is involved in AAA and its mechanism remains unknown. In this study, we found that the pyroptosis significantly increased in AAA tissues. ß5i inhibitor PR-957 treatment or ß5i deficiency markedly ameliorated AAA formation and decreased the pyroptosis. Pyroptosis were also significantly attenuated in bone marrow derived macrophages (BMDM) from ß5i-/- mice compared with the control group when they were subjected to OXLDL. Mechanistically, ß5i may promote activation of NFκB which augment NLRP3 expression. In conclusion, this study suggested macrophages pyroptosis are involved in AAA and inhibition or knockout of ß5i decreased macrophage pyroptosis via IκB/NFκB pathway.


Subject(s)
Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/pathology , Macrophages/immunology , Macrophages/pathology , Proteasome Endopeptidase Complex/immunology , Pyroptosis/immunology , Animals , Aortic Aneurysm, Abdominal/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Knockout, ApoE , NF-kappa B/metabolism , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/deficiency , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors/pharmacology , Pyroptosis/drug effects , Pyroptosis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Exp Lung Res ; 46(6): 174-184, 2020.
Article in English | MEDLINE | ID: mdl-32362153

ABSTRACT

Background: Previous studies have revealed the important role of alveolar macrophages (AMs) in the pathogenesis of acute respiratory distress syndrome (ARDS) and potential anti-inflammatory properties of lincRNA-p21. This study aims to study the association between lincRNA-p21 and active AMs to understand the molecular mechanisms of AMs-mediated inflammatory responses in ARDS.Methods: This study was mainly investigated in mice with the intratracheal instillation of lipopolysaccharide (LPS) or LPS-treated AMs. The expression of lincRNA-p21 and classical macrophage markers, IL-12ß and iNOS, was detected by quantitative RT-PCR, while NF-κB p65 translocation was measured by western blotting analysis. And, NF-κB activity was analyzed through luciferase report assays. Gain- and loss-of-function studies were also performed for further investigations.Results: Elevated lincRNA-p21 levels were observed in both LPS-induced ARDS mice and LPS-treated AMs, with upregulated expression of IL-12ß and iNOS, namely M1 activation, and p65 nuclear translocation. Further in vitro studies showed that LPS-induced M1 activation could be counteracted by both lincRNA-p21 inhibition and inhibited NF-κB activation. Moreover, both p65 nuclear translocation and NF-κB activity were promoted by lincRNA-p21 overexpression, while lincRNA-p21 inhibition showed a negative effect on LPS-induced p65 nuclear translocation and increase of NF-κB activity. Additionally, LPS-induced lung injuries could be attenuated by lincRNA-p21 inhibition in vivo.Conclusion: This study revealed elevated lincRNA-p21 levels in LPS-induced ARDS and investigated the potential role of lincRNA-p21 in LPS-induced pro-inflammatory response via NF-κB/p65 mediated pathways, suggesting the potential application of lincRNA-p21 for ADRS therapy.


Subject(s)
Macrophage Activation/genetics , Macrophages, Alveolar/metabolism , NF-kappa B/genetics , RNA, Long Noncoding/genetics , Respiratory Distress Syndrome/genetics , p21-Activated Kinases/genetics , Animals , Gene Expression Regulation/genetics , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Lung Injury/genetics , Macrophage Activation/drug effects , Macrophages, Alveolar/drug effects , Male , Mice , Mice, Inbred C57BL , Signal Transduction/genetics , Trachea/drug effects , Trachea/metabolism , Transcription Factor RelA/genetics
12.
Heart Vessels ; 34(5): 875-882, 2019 May.
Article in English | MEDLINE | ID: mdl-30535755

ABSTRACT

Inflammation plays a critical role in the development of abdominal aortic aneurysm (AAA). Chemokine receptor CXCR2 mediates inflammatory cell chemotaxis in several diseases. However, the role of CXCR2 in AAA and the underlying mechanisms remain unknown. In this study, we found that the CXCR2 expressions in AAA tissues from human and angiotensin II (Ang II)-infused apolipoprotein E knockout (Apo E-/-) mice were significantly increased. The pharmacological inhibition of CXCR2 (SB265610) markedly reduced Ang II-induced AAA formation. Furthermore, SB265610 treatment significantly reduced collagen deposition, elastin degradation, the metal matrix metalloprotease expression and accumulation of macrophage cells. In conclusion, these results showed CXCR2 plays a pathogenic role in AAA formation. Inhibition of CXCR2 pathway may represent a novel therapeutic approach to treat AAA.


Subject(s)
Aortic Aneurysm, Abdominal/drug therapy , Phenylurea Compounds/pharmacology , Receptors, Interleukin-8B/antagonists & inhibitors , Triazoles/pharmacology , Angiotensin II/adverse effects , Animals , Aortic Aneurysm, Abdominal/chemically induced , Disease Models, Animal , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout, ApoE , Signal Transduction
13.
Cell Physiol Biochem ; 42(4): 1550-1558, 2017.
Article in English | MEDLINE | ID: mdl-28723692

ABSTRACT

BACKGROUND: Activation of dendritic cells (DCs) is necessary to initiate immune responses. Angiotensin II (Ang II) has been reported to have a proinflammatory and immunomodulatory function. However, the role of Ang II in regulation of DCs and the underlying mechanisms remain illdefined. METHODS: The effects of Ang II on the proliferation, maturation, phagocytosis, migration, and communication with T cells of DCs were analysed utilizing MTT, flow cytometry, ELISA, transwell assay and mixed lymphocyte culture. RESULTS: We found that Ang II treatment significantly inhibited proliferation and phagocytic activity of DCs, but promoted the DC maturation and migration well as the expression of pro-inflammatory cytokines by DCs. In addition, Ang II also stimulated DC-mediated T cell proliferation. These effects were associated with activation of p65/NF-κB, ERK1/2 and STAT1 signaling pathways in DCs. CONCLUSIONS: Our results demonstrate that Ang II activates DCs partially through p65/NF-κB, ERK1/2 and STAT1 pathways, and suggest a potential therapeutic target of DC-mediated inflammatory disorders.


Subject(s)
Angiotensin II/pharmacology , Dendritic Cells/drug effects , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , STAT1 Transcription Factor/genetics , T-Lymphocytes/drug effects , Transcription Factor RelA/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Differentiation/drug effects , Cell Movement/drug effects , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/immunology , Gene Expression Regulation , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/immunology , Phagocytosis/drug effects , Primary Cell Culture , STAT1 Transcription Factor/immunology , Signal Transduction , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transcription Factor RelA/immunology
14.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 39(1): 120-127, 2017 Feb 20.
Article in English | MEDLINE | ID: mdl-28270294

ABSTRACT

Objective To investigate the role of proteasome inhibitor bortezomib (BTZ) in inflammatory response in abdominal aortic aneurysm (AAA) formation induced by angiotensin Ⅱ (Ang Ⅱ). Methods Ang Ⅱ-induced ApoE-/- mice AAA models were established. Forty male ApoE-/- mice (8-10-week-old) were randomly and equally divided into four groups:Sham group,BTZ group,Ang Ⅱ group,and Ang Ⅱ+BTZ group.HE staining,immunohistochemical staining,and flow cytometry were used to analyze the inflammatory response. Real-time quantitative polymerase chain reaction (qPCR) was used to analyze the mRNA expression of intercellular cell adhesion molecule-1 (ICAM-1). Western blotting was used to analyze the activation of nuclear factor κB signaling (NF-κB). Results The mean maximum suprarenal aortic diameter (Dmax) of Sham group,BTZ group,Ang Ⅱ group,and Ang Ⅱ+BTZ group were (1.00±0.01),(0.99±0.01),(1.50±0.13),and (1.20±0.04)mm,respectively (F=8.959,P=0.000). The Dmax of Ang Ⅱ group was significantly larger than those of Sham group (P=0.000) and Ang Ⅱ+BTZ group (P=0.015). The incidence of AAA in Ang Ⅱ group,Ang Ⅱ+BTZ group,and Sham group were 60%,17%,and 0,respectively. HE staining revealed that the abdominal aortic wall thickening was more severe in Ang Ⅱ group than in Sham group and Ang Ⅱ+BTZ group,similar with the infiltration of inflammatory cells. Immunohistochemical staining demonstrated that the CD3+T lymphocyte count was significantly higher in Ang Ⅱ group than in Sham group (107.9±15.9 vs. 0,P=0.000) and Ang Ⅱ+BTZ group (107.9±15.9 vs. 0.8±0.5,P=0.000). Flow cytometry also demonstrated that the proportion of the CD3+T lymphocytes of the Ang Ⅱ group [(13.50±0.69)%] was significantly higher than that in the Ang Ⅱ+BTZ group [(10.40±0.78)%] at week 1 (t=3.009,P=0.040),and the proportion of the CD3+T lymphocytes of the Ang Ⅱ group [(22.70±0.93)%] was significantly higher than that in the Ang Ⅱ+BTZ group [(15.10±0.97)%] at week 4 (t=5.654,P=0.005). The qPCR analysis showed that the mRNA expression of ICAM-1 was significantly up-regulated in Ang Ⅱ group than in Sham group (1.93±0.54 vs. 1.00±0.15,P=0.011) and Ang Ⅱ+BTZ group (1.93±0.54 vs. 0.83±0.08,P=0.009). Western blot analysis showed a lower phosphorylation level of inhibitor of NF-κB in the Ang Ⅱ group compared with the Sham group or Ang Ⅱ+BTZ group,accompanied with an increased phosphorylation level of p65. Conclusion Proteasome inhibitor BTZ can attenuate AAA formation partially by regulating T lymphocytes infiltration through regulating the mRNA expression of ICAM-1 regulated by the activation of NF-κB signaling pathway.


Subject(s)
Angiotensin II/adverse effects , Aortic Aneurysm, Abdominal/drug therapy , Bortezomib/pharmacology , Proteasome Inhibitors/pharmacology , Animals , Aortic Aneurysm, Abdominal/chemically induced , Apolipoproteins E/genetics , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Phosphorylation , Random Allocation , Signal Transduction , T-Lymphocytes/cytology
15.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 39(2): 188-195, 2017 Apr 20.
Article in English | MEDLINE | ID: mdl-28483016

ABSTRACT

Objective To investigated the changes of angiopoietin-like protein 2(Angptl2) in patients with arteriosclerotic occlusion (ASO). Methods A total of 140 subjects including 75 ASO patients (ASO group) and 65 healthy subjects (control group) were enrolled in this study. Angptl2 and adiponectin were evaluated by using enzyme-linked immunosorbent assay. Biochemical data and high sensitive C reactive protein were measured and recorded as well. Results Compared to the control group,the ASO group presented with significantly higher level of plasma Angptl2 [(13.55±9.17) µg/L vs. (9.04±4.79) µg/L,P=0.010]. Plasma Angptl2 level of critical limb ischemia subjects was significantly higher than that of intermittent claudication subjects [(17.01±10.20)µg/L vs. (10.53±6.97) µg/L,P=0.003]. The best diagnostic cutoff value of Angptl2 was 13.67 µg/L,with a sensitivity of 60.34% and a specificity of 81.25%. In addition,type 2 diabetes mellitus patients with ASO exhibited significantly higher serum Angptl2 levels [(18.67±9.84)µg/L] than those without ASO [(13.01±3.47) µg/L] (P=0.021). In ASO group,serum Angptl2 levels were negatively correlated with ankle brachial index (r=-0.244,P=0.035). Conclusion The plasma level of Angptl2 increases in ASO patients. Its level is remarkably increased when the disease progressions to critical limb ischemia. Angptl2 can be a potential biological marker of disease progression.


Subject(s)
Angiopoietin-like Proteins/blood , Arteriosclerosis/blood , Angiopoietin-Like Protein 2 , Biomarkers/blood , C-Reactive Protein/analysis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Humans
17.
Cell Biol Int ; 40(7): 742-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26916957

ABSTRACT

Airway smooth muscle cell (ASMC) was known to involve in the pathophysiology of asthma. Schisandrin B was reported to have anti-asthmatic effects in a murine asthma model. However, the molecular mechanism involving in the effect of Schisandrin B on ASMCs remains poorly understood. Sprague-Dawley rats were divided into three groups: rats as the control (Group 1), sensitized rats (Group 2), sensitized rats and intragastric-administrated Schisandrin B (Group 3). The expression of miR-135a and TRPC1 was detected in the rats from three groups. Platelet-derived growth factor (PDGF)-BB was used to induce the proliferation of isolated ASMCs, and the expression of miR-135a and TRPC1 was detected in PDGF-BB-treated ASMCs. Cell viability was examined in ASMCs transfected with miR-135a inhibitor or si-TRPC1. The expression of TRPC1 was examined in A10 cells pretreated with miR-135a inhibitor or miR-135a mimic. In this study, we found that Schisandrin B attenuated the inspiratory and expiratory resistances in sensitized rats. Schisandrin B upregulated the mRNA level of miR-135a and decreased the expression of TRPC1 in sensitized rats. In addition, Schisandrin B reversed the expression of miR-135a and TRPC1 in PDGF-BB-induced ASMCs. Si-TRPC1 abrogated the increasing proliferation of ASMCs induced by miR-135a inhibitor. We also found that miR-135a regulated the expression of TRPC1 in the A10 cells. These results demonstrate that Schisandrin B inhibits the proliferation of ASMCs via miR-135a suppressing the expression of TRPC1.


Subject(s)
Lignans/pharmacology , MicroRNAs/metabolism , Myocytes, Smooth Muscle/drug effects , Polycyclic Compounds/pharmacology , TRPC Cation Channels/biosynthesis , Airway Remodeling/drug effects , Animals , Apoptosis/drug effects , Asthma/drug therapy , Asthma/metabolism , Asthma/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cyclooctanes/pharmacology , Male , MicroRNAs/genetics , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Up-Regulation
18.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 38(2): 150-4, 2016 Apr.
Article in Zh | MEDLINE | ID: mdl-27181890

ABSTRACT

OBJECTIVE: To investigate the changes and value of plasma angiopoietin-related growth factor (AGF) in patients with abdominal aortic aneurysm (AAA). METHODS: Serum AGF level was analyzed in 50 AAA patients and in 56 healthy subjects. AGF and adiponectin were quantified by enzyme-linked immunosorbent assay. Routine testing of blood biochemistry and high-sensitivity C-reactive protein were performed. RESULTS: The plasma AGF level was significantly higher in AAA patients than in the controls [(87.91±96.87) µg/L vs. (56.89±41.32) µg/L, P=0.040],while serum adiponectin level showed no significant difference between these two groups. The plasma AGF level in patients with an AAA>5 cm and those with AAA between 3 cm and 5 cm were (96.08±68.61) µg/L and (75.27±46.05) µg/L. CONCLUSIONS: Plasma AGF is highly expressed in AAA patients. Higher serum AGF level is associates with larger AAA. Thus, AGF may be a potential serum biomarker for AAA.


Subject(s)
Angiopoietins/blood , Aortic Aneurysm, Abdominal/blood , Adiponectin/blood , Angiopoietin-Like Protein 6 , Angiopoietin-like Proteins , Biomarkers/blood , C-Reactive Protein/analysis , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Humans
19.
Apoptosis ; 20(4): 512-22, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25608996

ABSTRACT

NOD1 is a member of nucleotide-binding oligomerization domain-like receptors family that participates in many inflammatory processes. Previous studies demonstrated that NOD1 plays an important role in inflammatory cardiovascular diseases. However, its role in myocardial ischemia/reperfusion (I/R) injury remains unknown. The present study investigate whether NOD1 is involved in the pathogenesis of mouse myocardial I/R injury and the underlying mechanisms. Administration of NOD1 ligand (DAP) significantly enhanced myocardial I/R injury, as demonstrated by increased infarct size, the number of TUNEL-positive nuclei, caspase-3 activity, the infiltration of Mac-2- and IL-6-positive cells as compared with untreated heart or cardiomyocytes after I/R injury. In contrast, knockdown of NOD1 by siRNA markedly attenuated mimetic I/R induced cardiomyocyte apoptosis in vitro, indicating that NOD1 enhanced myocardial I/R injury partially through direct heart effects. These effects were partially associated with activation of JNK, p38 MAPK and NF-κB signaling pathways. Taken together, these results provide the first evidence that activation of intracellular sensor NOD1 enhances myocardial I/R injury and may provide novel therapeutic target for ameliorating the ischemic heart diseases.


Subject(s)
Diaminopimelic Acid/metabolism , Myocardial Ischemia/surgery , Myocardial Reperfusion Injury/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Signal Transduction , Animals , Apoptosis , Humans , Ligands , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/genetics , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/genetics
20.
Cell Physiol Biochem ; 35(2): 689-98, 2015.
Article in English | MEDLINE | ID: mdl-25591955

ABSTRACT

BACKGROUND/AIMS: Angiotensin II (Ang II) plays a critical role in regulating vascular inflammatory diseases, such as atherosclerosis and hypertension. Early growth response-1 (Egr-1) is an immediate early gene that acts as a master switch for the inflammatory response. However, the role of Ang II in regulating Egr-1 expression in macrophages, and the effect of peroxisome proliferators-activated receptor-γ (PPAR-γ) ligand 15d-PGJ2 in this process remain to be investigated. METHODS AND RESULTS: We showed that Ang II significantly up-regulated the expression of Egr-1 in RAW264.7 macrophages, and this effect was markedly attenuated by Eprosartan (an AT1R blocker), SP600125 (a JNK-specific inhibitor) and PD98059 (an ERK-specific inhibitor). Moreover, treatment of macrophages with 15d-PGJ2, a natural PPAR-γ ligand, significantly reduced Ang II-induced expression of Egr-1 and its inflammatory gene targets (IL-1ß, TNF-α, TGF-ß, MCP-1 and ICAM-1) through PPAR-γ activation and ROS formation. In addition, 15d-PGJ2 treatment markedly inhibited Ang II-induced macrophage migration and proliferation. CONCLUSIONS: This study for the first time demonstrates that Ang II can induce the expression of Egr-1 via AT1R/JNK and ERK signaling pathways. Activation of PPAR-γ by 15d-PGJ2 suppresses Egr-1-mediated proinflammatory response, and may be a novel therapeutic strategy for treatment of vascular inflammatory diseases.


Subject(s)
Angiotensin II/metabolism , Cytokines/metabolism , Early Growth Response Protein 1/metabolism , Macrophages/drug effects , Prostaglandin D2/analogs & derivatives , Acrylates/pharmacology , Animals , Anthracenes/pharmacology , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Prostaglandin D2/pharmacology , Thiophenes/pharmacology , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL