Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Lab ; 66(8)2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32776757

ABSTRACT

BACKGROUND: The present study focused on the potential clinical significance of Th-17 cell related inflammatory cytokines in the occurrence and development of neonatal respiratory distress syndrome (NRDS). METHODS: We included 82 NRDS children and 82 healthy controls. NRDS children were divided into the mild and severe group based on the disease severity. The serum samples of the NRDS and non-NRDS children were collected, and the expression levels of IL-17, IL-22, and IL-23 were determined by ELISA method. Moreover, correlation between the levels of the cytokines and the disease severity were analyzed, and receiver operating characteristics curve (ROC) analysis was performed to determine the diagnostic value of the cytokines. Finally, correlation between the lung ultrasound score (LUS) of the NRDS patients and the levels of IL-17 and IL-23 were analyzed. RESULTS: IL-17 and IL-23 were dramatically increased in serum of the NRDS patients compared with the non-NRDS patients; moreover, IL-17 and IL-23 were significantly higher in the severe compared with the mild NRDS group, and the levels of both IL-17 and IL-23 were positively correlated with the disease severity. Furthermore, ROC analysis showed that both IL-17 and IL-23 can distinguish NRDS patient, especially the severe NRDS patients from the non-NRDS patients with high sensitivity and specificity; finally, the levels of IL-17 and IL-23 were positively correlated with the LUS in NRDS patients. CONCLUSIONS: IL-17 and IL-23 were up-regulated in NRDS and may serve as sensitive biomarkers for the diagnosis and treatment of the disease.


Subject(s)
Interleukin-17 , Respiratory Distress Syndrome, Newborn , Child , Humans , Infant, Newborn , Interleukin-23 , Lung , ROC Curve
2.
J Cell Biochem ; 119(10): 8290-8303, 2018 11.
Article in English | MEDLINE | ID: mdl-29923351

ABSTRACT

Cardiac hypertrophy is one of the major risk factors for chronic heart failure. The role of endophilinA2 (EndoA2) in clathrin-mediated endocytosis and clathrin-independent endocytosis is well documented. In the present study, we tested the hypothesis that EndoA2 protects against angiotensin II (Ang II)-induced cardiac hypertrophy by mediating intracellular angiotensin II type 1 receptor (AT1-R) trafficking in neonatal rat cardiomyocytes (NRCMs). Cardiac hypertrophy was evaluated by using cell surface area and quantitative RT-PCR (qPCR) analyses. For the first time, we found that EndoA2 attenuated cardiac hypertrophy and fibrosis induced by Ang II. Moreover, EndoA2 inhibited apoptosis induced by excessive endoplasmic reticulum stress (ERS), which accounted for the beneficial effects of EndoA2 on cardiac hypertrophy. We further revealed that there was an interaction between EndoA2 and AT1-R.The expression levels of EndoA2, which inhibits AT1-R transport from the cytoplasm to the membrane, and the interaction between EndoA2 and AT1-R were obviously decreased after Ang II treatment. Furthermore, Ang II inhibited the co-localization of AT1-R with GRP-78, which was reversed by EndoA2 overexpression. In conclusion, our results suggested that EndoA2 plays a role in protecting against cardiac hypertrophy induced by Ang II, possibly by inhibiting AT1-R transport from the cytoplasm to the membrane to suppress signal transduction.


Subject(s)
Acyltransferases/genetics , Angiotensin II/genetics , Cardiomegaly/prevention & control , Myocytes, Cardiac/metabolism , Receptor, Angiotensin, Type 1/genetics , Acyltransferases/antagonists & inhibitors , Acyltransferases/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Animals, Newborn , Apoptosis/genetics , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Primary Cell Culture , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction , Transfection
3.
Cardiovasc Res ; 100(1): 134-42, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23786998

ABSTRACT

AIMS: We have previously demonstrated that ClC-3 chloride channel activity and expression are significantly increased in remodelled cerebral vessels of hypertensive rats. This study aims to examine whether this channel directly regulates cerebrovascular remodelling during hypertension by using ClC-3(-/-) mice. METHODS AND RESULTS: After DOCA-salt treatment, medial cross-sectional area, media thickness, and media-lumen ratio of the basilar artery of ClC-3(+/+) mice were significantly increased, accompanied by reduced lumen diameter, indicating apparent vascular remodelling. The vascular ultrastructure of ClC-3(+/+) hypertensive mice by electron microscopy revealed obvious disarray of SMCs and extracellular matrix accumulation. Immunofluorescence analysis showed that fibronectin was overexpressed in ClC-3(+/+) DOCA-salt mice. All of these vascular structure alterations were prevented in ClC-3(-/-) mice despite DOCA-salt treatment. However, propranolol, which reduced blood pressure as effectively as ClC-3 deficiency, failed to prevent basilar artery from remodelling. The vascular structure injury in ClC-3(+/+) hypertensive mice was accompanied by significantly increased expression of matrix metalloproteinase (MMP)-2, membrane-type (MT)1-MMP, and tissue inhibitor of metalloproteinase (TIMP)-2, which was inhibited by ClC-3 knockout. Additionally, the increase in transforming growth factor (TGF)-ß1 level in serum, as well as phosphorylation of Smad3 at serine 423/425 in basilar artery, induced by DOCA-salt, was markedly prevented in ClC-3(-/-) mice. CONCLUSION: Our findings suggest that ClC-3 deficiency attenuates cerebrovascular remodelling possibly via the suppression of MMPs/TIMP expression and TGF-ß1/Smad3 signalling pathway in this hypertension.


Subject(s)
Basilar Artery/pathology , Chloride Channels/physiology , Hypertension/pathology , Animals , Blood Pressure , Body Weight , Chloride Channels/deficiency , Desoxycorticosterone Acetate , Fibronectins/analysis , Hypertension/metabolism , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 9/analysis , Mice , Mice, Inbred C57BL , Phosphorylation , Smad3 Protein/physiology , Transforming Growth Factor beta1/blood , Transforming Growth Factor beta1/physiology
4.
Circulation ; 125(5): 697-707, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22215857

ABSTRACT

BACKGROUND: The Ca(2+)-activated chloride channel (CaCC) plays an important role in a variety of physiological functions. In vascular smooth muscle cells, CaCC is involved in the regulation of agonist-stimulated contraction and myogenic tone. The physiological functions of CaCC in blood vessels are not fully revealed because of the lack of specific channel blockers and the uncertainty concerning its molecular identity. METHODS AND RESULTS: Whole-cell patch-clamp studies showed that knockdown of TMEM16A but not bestrophin-3 attenuated CaCC currents in rat basilar smooth muscle cells. The activity of CaCC in basilar smooth muscle cells isolated from 2-kidney, 2-clip renohypertensive rats was decreased, and CaCC activity was negatively correlated with blood pressure (n=25; P<0.0001) and medial cross-sectional area (n=24; P<0.0001) in basilar artery during hypertension. Both upregulation of CaMKII activity and downregulation of TMEM16A expression contributed to the reduction of CaCC in the hypertensive basilar artery. Western blot results demonstrated that angiotensin II repressed TMEM16A expression in basilar smooth muscle cells (n=6; P<0.01). Knockdown of TMEM16A facilitated and overexpression of TMEM16A inhibited angiotensin II-induced cell cycle transition and cell proliferation determined by flow cytometry and BrdU incorporation (n=6 in each group; P<0.05). TMEM16A affected cell cycle progression mainly through regulating the expression of cyclin D1 and cyclin E. CONCLUSIONS: TMEM16A CaCC is a negative regulator of cell proliferation. Downregulation of CaCC may play an important role in hypertension-induced cerebrovascular remodeling, suggesting that modification of the activity of CaCC may be a novel therapeutic strategy for hypertension-associated cardiovascular diseases such as stroke.


Subject(s)
Basilar Artery/pathology , Cell Proliferation , Chloride Channels/metabolism , Down-Regulation , Hypertension/pathology , Muscle, Smooth, Vascular/pathology , Animals , Anoctamin-1 , Basilar Artery/metabolism , Bestrophins , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Cycle/physiology , Cells, Cultured , Disease Models, Animal , Hypertension/metabolism , Male , Muscle, Smooth, Vascular/metabolism , Patch-Clamp Techniques , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...