Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 207: 115350, 2023 01.
Article in English | MEDLINE | ID: mdl-36435201

ABSTRACT

The pulmonary vascular remodeling (PVR), the pathological basis of pulmonary hypertension (PH), entails pulmonary artery smooth muscle cells (PASMCs) phenotypic switching, but appreciation of the underlying mechanisms is incomplete. Exosomes, a novel transfer machinery enabling delivery of its cargos to recipient cells, have been recently implicated in cardiovascular diseases including PH. The two critical questions of whether plasma-derived exosomes drive PASMCs phenotypic switching and what cargo the exosomes transport, however, remain unclear. Herein, by means of transmission electron microscopy and protein detection, we for the first time, characterized lectin like oxidized low-density lipoprotein receptor-1 (LOX-1) as a novel cargo of plasma-derived exosomes in PH. With LOX-1 knockout (Olr1-/-) rats-derived exosomes, we demonstrated that exosomal LOX-1 could be transferred into PASMCs and thus elicited cell phenotypic switching. Of importance, Olr1-/- rats exhibited no cell phenotypic switching and developed less severe PH, but administration of wild type rather than Olr1-/- exosomes to Olr1-/- rats recapitulated the phenotype of PH with robust PASMCs phenotypic switching. We also revealed that exosomal LOX-1 triggered PASMCs phenotypic switching, PVR and ultimately PH via ERK1/2-KLF4 signaling axis. This study has generated proof that plasma-derived exosomes confer PH by delivering LOX-1 into PASMCs. Hence, exosomal LOX-1 represents a novel exploitable target for PH prevention and treatment.


Subject(s)
Exosomes , Hypertension, Pulmonary , Rats , Animals , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Hypertension, Pulmonary/metabolism , Exosomes/metabolism , Cell Proliferation/physiology , Hypoxia/metabolism , Phenotype , Myocytes, Smooth Muscle/metabolism , Scavenger Receptors, Class E/genetics , Scavenger Receptors, Class E/metabolism , Cells, Cultured , Vascular Remodeling/physiology
2.
Mol Neurobiol ; 60(3): 1563-1579, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36527595

ABSTRACT

Glutamate receptor ionotropic NMDA 2B (GluN2B) plays an essential role in calcium overload during excitotoxicity. Reverse-phase nano-liquid chromatography-tandem mass spectrometry has revealed an interaction between GluN2B and HECT domain E3 ubiquitin protein ligase 4 (HECTD4), an E3 ubiquitin ligase highly expressed in the brain. As a potential substrate for HECTD4, mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) acts as a scaffold with hydrolysis activity. This study explores the relationship between HECTD4, GluN2B, and MALT1, focusing on their role in brain injury in ischemic stroke. Rats were subjected to 2 h-ischemia followed by 24-h reperfusion to establish an ischemic stroke model. We observed the downregulation of HECTD4 and the upregulation of MALT1. Additionally, an increased GluN2B phosphorylation was concomitant with weakened interactions between HECTD4 and GluN2B, followed by decreased striatal-enriched protein phosphatase (STEP61). Knockdown of HECTD4 exacerbated hypoxia- or NMDA-induced injury in nerve cells coincident with a decrease in GluN2B and MALT1 ubiquitination, and an increase in GluN2B phosphorylation as well as an increase in intracellular calcium level, which were counteracted by MALT1 siRNA. Blockage of MALT1 with its inhibitor or siRNA reduced STEP61 degradation, accompanied by a decrease in GluN2B phosphorylation, intracellular calcium concentration, and brain cell injury, which were reversed by overexpression of MALT1. Based on these observations, we conclude that the downregulation of HECTD4 in ischemic stroke rat brain accounts for calcium overload and brain injury due to activating GluN2B directly and indirectly through a mechanism involving the reduced ubiquitination of GluN2B and MALT1, respectively.


Subject(s)
Brain Injuries , Ischemic Stroke , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Receptors, N-Methyl-D-Aspartate , Ubiquitin-Protein Ligases , Animals , Rats , Brain Injuries/complications , Calcium , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , N-Methylaspartate , Receptors, N-Methyl-D-Aspartate/metabolism , RNA, Small Interfering , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
3.
J Cardiovasc Transl Res ; 15(5): 1129-1142, 2022 10.
Article in English | MEDLINE | ID: mdl-35239171

ABSTRACT

Endosomal sorting complex required for transport III (ESCRT-III) machinery is a key component to counteract the mixed lineage kinase domain-like pseudokinase (MLKL)-induced plasma membrane broken in cells undergoing necroptosis. Based on the bioinformatics analysis, polymyxin B, a polypeptide antibiotic, is predicted to simultaneously interact with ESCRT-III subunits and necroptosis-relevant proteins. This study aims to explore whether polymyxin B could reduce necroptosis in the stroke rat brain via enhancing the ESCRT-III machinery and/or suppressing the RIPK1/RIPK3/MLKL pathway. The stroke rats showed evident brain injury, concomitant with the downregulation of ESCRT-III subunits and the upregulation of necroptosis-relevant proteins. Post-ischemic administration of polymyxin B could alleviate the brain injury, accompanied by restoration of the levels of ESCRT-III subunits and suppression of necroptosis-relevant proteins. And, polymyxin B exerted similar effects in hypoxia-treated HT22 cells. We conclude that polymyxin B can reduce necroptosis in the stroke rat brain via enhancing the ESCRT-III machinery and suppressing the RIPK1/RIPK3/MLKL pathway simultaneously.


Subject(s)
Brain Injuries , Ischemic Stroke , Stroke , Animals , Rats , Endosomal Sorting Complexes Required for Transport , Polymyxin B , Protein Kinases/metabolism
4.
Zhongguo Dang Dai Er Ke Za Zhi ; 24(2): 216-222, 2022 Feb 15.
Article in English, Chinese | MEDLINE | ID: mdl-35209989

ABSTRACT

Multisystem inflammatory syndrome in children (MIS-C) is a type of hyperinflammatory symptoms similar to Kawasaki disease after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and is commonly observed in children aged 8-10 years. Primary therapeutic medications for MIS-C are intravenous immunoglobulins and glucocorticoids. It has been reported that biologics, such as IL-1 receptor antagonist anakinra, IL-6 receptor antagonist tocilizumab, and TNF-α receptor antagonist infliximab, can be used as an option for critically ill patients. This article elaborates on the mechanism of action of the above biologics and discusses the efficacy and safety biologics in the treatment of MIS-C after SARS-CoV-2 infection, in order to provide methods for the treatment of MIS-C with severe symptoms.


Subject(s)
Biological Products , COVID-19 , COVID-19/complications , Child , Humans , SARS-CoV-2 , Systemic Inflammatory Response Syndrome
5.
Biomed Res Int ; 2021: 2202888, 2021.
Article in English | MEDLINE | ID: mdl-34513987

ABSTRACT

The proliferation of pulmonary artery smooth muscle cells (PASMCs) is an important cause of pulmonary vascular remodeling in pulmonary hypertension (PH). It has been reported that miR-137 inhibits the proliferation of tumor cells. However, whether miR-137 is involved in PH remains unclear. In this study, male Sprague-Dawley rats were subjected to 10% O2 for 3 weeks to establish PH, and rat primary PASMCs were treated with hypoxia (3% O2) for 48 h to induce cell proliferation. The effect of miR-137 on PASMC proliferation and calpain-2 expression was assessed by transfecting miR-137 mimic and inhibitor. The effect of calpain-2 on PASMC proliferation was assessed by transfecting calpain-2 siRNA. The present study found for the first time that miR-137 was downregulated in pulmonary arteries of hypoxic PH rats and in hypoxia-treated PASMCs. miR-137 mimic inhibited hypoxia-induced PASMC proliferation and upregulation of calpain-2 expression in PASMCs. Furthermore, miR-137 inhibitor induced the proliferation of PASMCs under normoxia, and knockdown of calpain-2 mRNA by siRNA significantly inhibited hypoxia-induced proliferation of PASMCs. Our study demonstrated that hypoxia-induced downregulation of miR-137 expression promoted the proliferation of PASMCs by targeting calpain-2, thereby potentially resulting in pulmonary vascular remodeling in hypoxic PH.


Subject(s)
Calpain/genetics , Hypertension, Pulmonary/genetics , MicroRNAs/genetics , Animals , Calpain/metabolism , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Hypertension, Pulmonary/pathology , Hypoxia/genetics , Hypoxia/metabolism , Male , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Pulmonary Artery/metabolism , Pulmonary Artery/physiology , Rats , Rats, Sprague-Dawley , Vascular Remodeling/genetics
6.
Eur J Pharmacol ; 908: 174307, 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34245748

ABSTRACT

Pulmonary vascular remodeling (PVR) is the pathological basis of pulmonary hypertension (PH). Incomplete understanding of PVR etiology has hindered drug development for this devastating disease, which exhibits poor prognosis despite the currently available therapies. Endothelial-to-mesenchymal transition (EndMT), a process of cell transdifferentiation, has been recently implicated in cardiovascular diseases, including PH. But the questions of how EndMT occurs and how to pharmacologically target EndMT in vivo have yet to be further answered. Herein, by performing hematoxylin-eosin and immunofluorescence staining, transmission electron microscopy and Western blotting, we found that EndMT plays a key role in the pathogenesis of PH, and importantly that aspirin, a FDA-approved widely used drug, was capable of ameliorating PVR in a preclinical rat model of hypoxia-induced PH. Moreover, aspirin exerted its inhibitory effects on EndMT in vitro and in vivo by suppressing HIF-1α/TGF-ß1/Smads/Snail signaling pathway. Our data suggest that EndMT represents an intriguing drug target for the prevention and treatment of hypoxic PH and that aspirin may be repurposed to meet the urgent therapeutic needs of hypoxic PH patients.


Subject(s)
Hypertension, Pulmonary , Vascular Remodeling , Animals , Aspirin/pharmacology , Endothelium , Epithelial-Mesenchymal Transition , Rats , Smad3 Protein , Transforming Growth Factor beta1
7.
Life Sci ; 255: 117822, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32450174

ABSTRACT

AIM: Proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) are regarded as the primary factors resulting in pulmonary arterial remodeling in pulmonary hypertension (PH). Myeloid ecotropic viral integration site 1 (MEIS1) has been positioned as a negative cardiomyocyte cell cycle regulator and regulates proliferation of multiple kinds of cancer cells. Whether MESI1 is involved in the proliferation and migration of PASMCs deserves to be identified. MAIN METHODS: Sprague Dawley rats were exposed to hypoxia condition (10% O2) for 4 weeks to induce PH and primary rat PASMCs were cultured in hypoxia condition (3% O2) for 48 h to induce proliferation and migration. Immunohistochemistry, immunofluorescence, reverse transcription PCR and Western blot analysis were performed to detect the expressions of target mRNAs and proteins. EDU, CCK8 and wound healing assays were conducted to measure the proliferation and migration of PASMCs. KEY FINDINGS: Hypoxia down-regulated the expression of MEIS1 (both mRNA and protein) in pulmonary arteries and PASMCs. Over-expression of MEIS1 inhibited the proliferation and migration of PASMCs afforded by hypoxia. In contrast, knockdown of MEIS1 under normoxia condition like hypoxia induced the proliferation and migration of PASMCs. MEIS1 mediated hypoxia-induced the proliferation and migration of PASMCs via METTL14/MEIS1/p21 signaling. SIGNIFICANCE: The present study revealed that MEIS1 regulated the proliferation and migration of PASMCs during hypoxia-induced PH. Thus, MEIS1 may be a potential target for PH therapy.


Subject(s)
Hypertension, Pulmonary/physiopathology , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myocytes, Smooth Muscle/cytology , Pulmonary Artery/cytology , Animals , Cell Movement/physiology , Cell Proliferation/physiology , Cells, Cultured , Hypertension, Pulmonary/genetics , Hypoxia , Male , Muscle, Smooth, Vascular/cytology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Vascular Remodeling/physiology
8.
Eur J Pharmacol ; 864: 172698, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31563647

ABSTRACT

It has well been demonstrated that E3 ubiquitin ligase cullin7 plays important roles in cancer cell growth control via down-regulating p53 expression. The noncanonical function or the pathogenic role of p53 has more recently been implicated in pulmonary vascular remodeling. Therefore, whether cullin7 participates in hypoxia-induced pulmonary vascular remodeling deserves to be elucidated. The present study found that hypoxia up-regulated the expression of cullin7 mRNA and protein in pulmonary arteries and pulmonary artery smooth muscle cells, and knockdown of cullin7 inhibited hypoxia-induced proliferation and migration of pulmonary artery smooth muscle cells and reversed hypoxia-induced inhibition of p53 expression. Notably, administration of proteasome inhibitor MG132 significantly inhibited the expression of cullin7 and up-regulated the expression of p53 in pulmonary arteries concomitantly with improvement of hypoxia-induced pulmonary vascular remodeling. Our study demonstrated that hypoxia induced up-regulation of cullin7 expression resulting to the proliferation and migration of pulmonary artery smooth muscle cells via down-regulating p53 expression, which contributed to pulmonary vascular remodeling.


Subject(s)
Cell Movement , Cullin Proteins/metabolism , Hypertension, Pulmonary/pathology , Hypoxia/complications , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/pathology , Up-Regulation , Animals , Cell Proliferation , Cullin Proteins/genetics , Gene Knockdown Techniques , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/metabolism , Leupeptins/pharmacology , Male , Myocytes, Smooth Muscle/drug effects , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , Vascular Remodeling/drug effects
9.
Eur J Pharmacol ; 863: 172673, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31542480

ABSTRACT

Pulmonary arterial remodeling is a crucial cause of increased pulmonary artery pressure during pulmonary hypertension (PH). Recently, growing evidence has upheld the contribution of endothelial-mesenchymal transition (EndMT) to pulmonary arterial remodeling, but the underlying mechanisms remain largely unaddressed. miR-204 has been implicated in PH, being anti-proliferative and pro-apoptotic in pulmonary artery smooth muscles cells (PASMCs), but its role in EndMT is still unknown. Here we found that miR-204 was down-regulated by hypoxia in rat pulmonary arterial intima and human pulmonary artery endothelial cells (HPAECs), and its further down-regulation by using miR-204 inhibitor suppressed hypoxia-induced EndMT. Moreover, autophagy, evoked by hypoxia in rat pulmonary arterial intima and HPAECs, suppressed hypoxia-induced EndMT via p62-dependent degradation of Snail and Twist. Additionally, autophagy was regulated by miR-204 targeting ATG7. While down-regulation of miR-204 in PASMCs reportedly promoted monocrotaline-induced pulmonary arterial hypertension via increased cell proliferation, our data suggested an important, albeit dichotomous, role of miR-204 down-regulation in endothelial cells in the process of EndMT that it attenuated EndMT by enhancing autophagy, thereby ameliorating hypoxia-induced PH to some extent.


Subject(s)
Autophagy/genetics , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Autophagy-Related Protein 7/genetics , Cell Hypoxia/genetics , Cell Line , Humans , Male , Proteolysis , Rats , Rats, Sprague-Dawley , Snail Family Transcription Factors/metabolism , Twist-Related Protein 1/metabolism
10.
Life Sci ; 227: 64-73, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-31004656

ABSTRACT

AIM: Growing evidence suggests that endothelial-mesenchymal transition (EndMT) play key roles in pulmonary arterial remodeling during pulmonary arterial hypertension (PAH), but the underlying mechanisms have yet to be fully understood. miR-27a has been shown to promote proliferation of pulmonary arterial cells during PAH, but its role in EndMT remains unexplored. This study was designed to investigate the role and underlying mechanism of miR-27a in EndMT during PAH. MAIN METHODS: Rats were exposed in hypoxia (10% O2) for 3 weeks to induce PAH, and human pulmonary artery endothelial cells (HPAECs) were exposed in hypoxia (1% O2) for 48 h to induce EndMT. Immunohistochemistry, in situ hybridization, immunofluorescence, real-time PCR and Western blot were conducted to detect the expressions of RNAs and proteins, and luciferase assay was used to verify the putative binding site of miR-27a. KEY FINDINGS: We found that hypoxia up-regulated miR-27a in the tunica intima of rat pulmonary arteries and HPAECs, and that inhibition of miR-27a suppressed hypoxia-induced EndMT. Furthermore, elevated expression of miR-27a suppressed bone morphogenetic protein (BMP) signaling by targeting Smad5, thereby lessening Id2-mediated repression of the 2 critical mediators of EndMT (Snail and Twist). SIGNIFICANCE: Our data unveiled a novel role of miR-27a in EndMT during hypoxia-induced PAH. Thus, targeting of miR-27a-related pathway may be therapeutically harnessed to treat PAH.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Hypertension, Pulmonary/genetics , MicroRNAs/physiology , Animals , Cell Culture Techniques , Cell Proliferation , Endothelial Cells/metabolism , Endothelium/metabolism , Gene Expression Regulation , Humans , Hypertension, Pulmonary/physiopathology , Hypoxia/genetics , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Pulmonary Artery/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation , Vascular Remodeling/genetics
11.
Clin Exp Hypertens ; 40(2): 192-201, 2018.
Article in English | MEDLINE | ID: mdl-28872356

ABSTRACT

BACKGROUND: We have previously reported that the long-term exposure of organophosphorus induces vascular dementia (VD) in rats. As a coenzyme, vitamin B6 is mainly involved in the regulation of metabolisms. Whether vitamin B6 improves VD remains unknown. METHODS: The model of VD was induced by feeding rats with isocarbophos (0.5 mg/kg per two day, 12 weeks). The blood flow of the posterior cerebral artery (PCA) in rat was assessed by transcranial Doppler (TCD). The learning and memory were evaluated by the Morris Water Maze (MWM) test. RESULTS: Administration of vitamin B6 increased the blood flow in the right and left posterior cerebral arteries and improved the functions of learning and memory in isocarbophos-treated rats. Vitamin B6 increased the protein levels of N-methyl-D-aspartate receptor (NMDAR) 2B, postsynaptic densities (PSDs) protein 95, and calmodulin-dependent protein kinase II (CaMK-II) in the hippocampus, which were decreased by isocarbophos in rats. Morphological analysis by light microscope and electronic microscope indicated disruptions of the hippocampus caused by isocarbophos were normalized by vitamin B6. Importantly, the antagonist of NMDAR signaling by eliprodil abolished these beneficial effects produced by vitamin B6 on PCA blood flow, learning, memory, and hippocampus structure in rats, as well as the protein expression of NMDAR 2B, PSDs protein 95, and CaMK-II in the hippocampus. CONCLUSION: Vitamin B6 activates NMDAR signaling to prevent isocarbophos-induced VD in rats.


Subject(s)
Dementia, Vascular/metabolism , Dementia, Vascular/prevention & control , Receptors, N-Methyl-D-Aspartate/metabolism , Vitamin B 6/pharmacology , Vitamin B Complex/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cerebrovascular Circulation/drug effects , Dementia, Vascular/chemically induced , Disks Large Homolog 4 Protein/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Hippocampus/ultrastructure , Hypertension/physiopathology , Malathion/analogs & derivatives , Male , Maze Learning/drug effects , Memory/drug effects , Piperidines/pharmacology , Posterior Cerebral Artery/diagnostic imaging , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Ultrasonography, Doppler
13.
Eur J Pharmacol ; 809: 42-51, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28478070

ABSTRACT

Pulmonary hypertension (PH) mainly results from excessive proliferation of pulmonary artery smooth muscle cells (PASMCs) and displays mitochondrial abnormalities such as mitochondrial fragmentation. Epigallocatechin-3-gallate (EGCG), an efficient antiproliferative compound in green tea, has recently been demonstrated to inhibit PASMCs proliferation. However, the pre-clinical issues as to whether EGCG attenuates PH and the underlying mechanisms have yet to be addressed. The present study was undertaken to investigate the therapeutic effects of EGCG on PH and its effects on mitochondrial fragmentation in PASMCs. Rats exposed to hypoxia (10% O2, 3 weeks) developed PH. EGCG (50, 100 or 200mg/kg/d, i.g.) dose-dependently attenuated right ventricular systolic pressure, pulmonary vascular remodeling and right ventricular hypertrophy, increased expression of mitochondrial fusion protein - mitofusin-2 (MFN-2), and promoted mitochondrial fusion as evidenced by decreased number and volume of mitochondria in PASMCs of pulmonary arteries. Notably, EGCG (50µM) downregulated hypoxia-induced (3% O2, 48h) PASMCs mitochondrial fragmentation and inhibited PASMCs proliferation via KLF-4/MFN-2/p-Erk signaling pathway. Collectively, our data demonstrated that EGCG exerts antiproliferative effects via regulating mitochondrial fragmentation of PASMCs and EGCG holds the promise as a drug against PH.


Subject(s)
Catechin/analogs & derivatives , Hypoxia/pathology , Membrane Proteins/metabolism , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Vascular Remodeling/drug effects , Animals , Catechin/pharmacology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , GTP Phosphohydrolases , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Up-Regulation/drug effects
14.
Life Sci ; 174: 35-42, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28259654

ABSTRACT

AIM: Chronic hypoxia leads to right ventricular hypertrophy (RVH). RVH is believed to result from hypoxia-induced pulmonary hypertension. However, if hypoxia impacts RVH directly awaits clarification. Hypoxia triggers oxidative stress, and lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) mediates reactive oxygen species (ROS) generation in different cells. Therefore, this study aims to explore whether LOX-1-mediated oxidative stress accounts for hypoxia-induced RVH. MAIN METHODS: Rats developed RVH after 3weeks of hypoxia (10% O2). Immunofluorescence staining was performed to evaluate H9C2 cell hypertrophy induced by hypoxia (3% O2). Real-time PCR and Western-blot were performed to assess LOX-1, NADPH oxidases (NOX), collagen I/III, atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) expression. DCFH-DA staining was performed to measure ROS generation. KEY FINDINGS: Hypoxia induced RVH and cardiac fibrosis in rats, as indicated by enlarged cardiomyocytes and deposition of extracellular matrix. Interestingly, hypoxia treatment directly induced H9C2 cardiomyocyte hypertrophy, implying direct effects of hypoxia on cell hypertrophy. Rat and H9C2 hypertrophy model revealed that cell hypertrophy was accompanied by marked increase in LOX-1 expression. Knockdown of LOX-1 significantly ameliorated H9C2 cell hypertrophy. Mechanistically, hypoxia induced prominent oxidative stress in rat right ventricles and H9C2 cells, most likely as a result from increased expression of NOX2/4, contributing to RVH. Knockdown of LOX-1 significantly attenuated H9C2 cell oxidative stress, with a concomitant decrease in NOX2/4 expression. SIGNIFICANCE: LOX-1/NOX/ROS pathway could represent a novel mechanism underlying hypoxia-induced RVH. Therapeutic targeting of LOX-1 would be exploited to treat RVH owing to chronic hypoxia exposure.


Subject(s)
Hypertrophy, Right Ventricular/etiology , Hypoxia/complications , Scavenger Receptors, Class E/metabolism , Animals , Blotting, Western , Cells, Cultured , Fluorescent Antibody Technique , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/pathology , Hypoxia/physiopathology , Male , Oxidative Stress/drug effects , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Scavenger Receptors, Class E/genetics , Signal Transduction/drug effects
15.
Pulm Pharmacol Ther ; 44: 70-77, 2017 06.
Article in English | MEDLINE | ID: mdl-28315789

ABSTRACT

BACKGROUND AND OBJECTIVE: Diabetic pulmonary fibrosis is a severe disease that increases mortality risk of diabetes. However, the molecular mechanisms leading to pulmonary fibrosis in diabetes are poorly understood. This study investigated the roles of epithelial-mesenchymal transition (EMT) and the associated molecular mechanisms in streptozotocin (STZ)-induced rat pulmonary fibrosis. METHODS: The rat model of diabetic pulmonary fibrosis was established by intraperitoneal injection of a single dose of STZ (35 mg/kg). Typical lesions of diabetic pulmonary fibrosis were observed 8 weeks after STZ injection by hematoxylin-eosin (HE) and Masson staining. Human bronchial epithelial cells (HBECs) and A549 cells were treated by high glucose. Gene or protein expression was measured by real-time PCR, Western blot, immunohistochemistry or immunofluorescence. The knockdown of lectin-like oxidized low density lipoprotein receptor-1 (LOX-1) or transforming growth factor-ß1 (TGF-ß1) was conducted by siRNA. RESULTS: Activation of EMT was observed in lung tissues of STZ-induced diabetic rats, exhibiting a loss in the epithelial cell marker E-cadherin and an increase in the mesenchymal marker Vimentin. The protein and mRNA levels of LOX-1, TGF-ß1 and krüppel-like factor 6 (KLF6) in the lung tissues were increased. Incubation of HBECs and A549 cells with high glucose activated EMT and induced an increase in LOX-1, TGF-ß1 and KLF-6 expression. LOX-1 siRNA inhibited high glucose-induced EMT in HBECs and A549 cells, which correlated with the reduction of TGF-ß1. TGF-ß1 siRNA decreased the expression of LOX-1 and KLF6. CONCLUSIONS: EMT was involved in the pathological process of diabetic pulmonary fibrosis, which was activated by LOX-1/TGF-ß1/KLF6 signaling pathway.


Subject(s)
Diabetes Mellitus, Experimental/complications , Epithelial-Mesenchymal Transition/physiology , Lung/pathology , Pulmonary Fibrosis/etiology , A549 Cells , Animals , Blotting, Western , Cadherins/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Kruppel-Like Factor 6/genetics , Kruppel-Like Factor 6/metabolism , Lung/metabolism , Rats , Real-Time Polymerase Chain Reaction , Scavenger Receptors, Class E/genetics , Scavenger Receptors, Class E/metabolism , Signal Transduction/physiology , Streptozocin , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Vimentin/metabolism
16.
Biochem Biophys Res Commun ; 488(4): 655-663, 2017 07 08.
Article in English | MEDLINE | ID: mdl-28108289

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) is a proliferative disorder associated with enhanced proliferation and suppressed apoptosis of pulmonary artery smooth muscle cells (PASMCs). Our lately study demonstrated that let-7g inhibited hypoxia-induced proliferation of PASMCs via repressing c-myc-Bmi-1-p16 signaling pathway. However, the upstream of let-7g has not yet been fully defined. Previous studies have shown that LOX-1, a target of let-7g, could also regulate the expression of let-7g in human aortic endothelial cells. In this present study, we aimed to investigate whether there is a negative feedback regulation between microRNA let-7g and LOX-1 in hypoxia-induced proliferation of PASMCs. METHODS: SD Rats were exposed to hypoxia (10% O2, 3 weeks) to induce PH. HE staining was used to evaluate pulmonary artery remodeling. in situ hybridization and immunohistochemistry were performed to assess the expression and distribution of let-7g and LOX-1, respectively. MTS, EDU and flow cytometry were performed to evaluate PASMCs proliferation. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were conducted to assess the expression of let-7g, LOX-1, calpain-1,-2,-4, and OCT-1. RESULTS: The expression of let-7g was significantly down-regulated in pulmonary arteries of hypoxia-induced PH rats accompanied by pulmonary vascular remodeling, whereas let-7g mimic inhibited hypoxia-induced proliferation of PASMCs and up-regulation of LOX-1 expression. LOX-1 blocking reversed hypoxia-induced down-regulation of let-7g expression. Calpains, protein kinase C and OCT-1 were involved in negative feedback regulation between let-7g and LOX-1. CONCLUSION: Negative feedback regulation between let-7g and LOX-1 mediated hypoxia-induced proliferation of in PASMCs.


Subject(s)
Feedback, Physiological , Hypoxia , MicroRNAs/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/cytology , Scavenger Receptors, Class E/metabolism , Animals , Cell Proliferation , Down-Regulation , Male , MicroRNAs/genetics , Rats , Rats, Sprague-Dawley , Scavenger Receptors, Class E/genetics
17.
Eur J Pharmacol ; 796: 190-206, 2017 Feb 05.
Article in English | MEDLINE | ID: mdl-27916556

ABSTRACT

MicroRNAs (miRNAs), a large family of small and highly conserved non-coding RNAs, regulate gene expression through translational repression or mRNA degradation. Aberrant expression of miRNAs underlies a spectrum of diseases including organ fibrosis. Recent evidence suggests that miRNAs contribute to organ fibrosis through mediating epithelial-mesenchymal transition (EMT). Alleviation of EMT has been proposed as a promising strategy against fibrotic diseases given the key role of EMT in fibrosis. miRNAs impact the expression of specific ligands, receptors, and signaling pathways, thus modulating EMT and consequently influencing fibrosis. This review summarizes the current knowledge concerning how miRNAs regulate EMT and highlights the specific roles that miRNAs-regulated EMT plays in fibrotic diseases as diverse as pulmonary fibrosis, hepatic fibrosis, renal fibrosis and cardiac fibrosis. It is desirable that a more comprehensive understanding of the functions of miRNAs-regulated EMT will facilitate the development of novel diagnostic and therapeutic strategies for various debilitating organ fibrosis.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Fibrosis/genetics , Fibrosis/pathology , MicroRNAs/genetics , Animals , Humans
18.
Can J Physiol Pharmacol ; 94(12): 1315-1324, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27556497

ABSTRACT

We have found that eIF3a plays an important role in bleomycin-induced pulmonary fibrosis, and up-regulation of eIF3a induced by TGF-ß1 is mediated via the ERK1/2 pathway. Whether ERK1/2 - eIF3a signal pathway is involved in calcitonin gene-related peptide (CGRP)-mediated pathogenesis of bleomycin-induced pulmonary fibrosis remains unknown. Pulmonary fibrosis was induced by intratracheal instillation of bleomycin (5 mg/kg) in rats. Primary pulmonary fibroblasts were cultured to investigate the proliferation by BrdU incorporation method and flow cytometry. Sensory CGRP depletion by capsaicin exacerbated bleomycin-induced pulmonary fibrosis in rats, as shown by a significant disturbed alveolar structure, marked thickening of the interalveolar septa and dense interstitial infiltration by inflammatory cells and fibroblasts, accompanied with increased expression of TGF-ß1, eIF3a, phosphorylated ERK1/2, α-SMA, collagen I, and collagen III. Exogenous application of CGRP significantly inhibited TGF-ß1-induced proliferation and differentiation of pulmonary fibroblasts concomitantly with decreased expression of eIF3a, phosphorylated ERK1/2, α-SMA, collagen I, and collagen III. These effects of CGRP were abolished in the presence of CGRP8-37. These results suggest that endogenous CGRP is related to the development of pulmonary fibrosis induced by bleomycin, and the inhibitory effect of CGRP on proliferation of lung fibroblasts involves the ERK1/2 - eIF3a signaling pathway.


Subject(s)
Bleomycin/toxicity , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/pharmacology , Down-Regulation/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Calcitonin Gene-Related Peptide/therapeutic use , Cells, Cultured , Down-Regulation/physiology , Fibroblasts/drug effects , Fibroblasts/metabolism , Male , Pulmonary Fibrosis/drug therapy , Random Allocation , Rats , Rats, Sprague-Dawley
20.
Biochem Biophys Res Commun ; 471(4): 402-8, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26906623

ABSTRACT

BACKGROUND: Endothelial cell apoptosis contributes to cardiovascular diseases such as hypertension, atherosclerosis. MicroRNA regulates endothelial cell function but its role in endothelial cell apoptosis remains to be fully elucidated. This study aims to investigate the role of miR-590-5p in endothelial cell apoptosis and dissect the underlying mechanisms. METHODS: Flow cytometric analysis, Hoechst 33258 staining and Western blotting were performed to evaluate human umbilical vein endothelial cell (HUVEC) apoptosis induced by Angiotensin (Ang) II. Western blotting and real-time quantitative PCR were conducted to assess the expression of LOX-1. DCFH-DA staining was carried out to measure the generation of reactive oxygen species (ROS). RESULTS: Ang II-induced HUVEC apoptosis was accompanied by downregulation of miR-590-5p; administration of miR-590-5p mimics attenuated HUVEC apoptosis and decreased ROS generation, as indicated by reduced fraction of apoptotic HUVECs and decreased caspase-3 activity. LOX-1 expression was increased by Ang II, and miR-590-5p mimics reduced LOX-1 expression in HUVECs in the absence or presence of Ang II. Pharmacologic or genetic block of LOX-1 with small interference RNA or TS92 (LOX-1 neutralizing antibody) significantly ameliorated HUVEC apoptosis, as evidenced by reduced number of apoptotic HUVECs, inhibited caspase-3 activation and suppressed mitochondrial cytochrome C release. Moreover, LOX-1 siRNA or TS92 treatment dramatically reduced ROS production in HUVECs treated with Ang II. CONCLUSION: Our data demonstrated that miR-590-5p downregulation promoted Ang II-induced endothelial cell apoptosis by elevating LOX-1 expression and consequently increasing ROS generation. Thus, restoration of miR-590-5p or block of LOX-1 could be therapeutically exploited to alleviate endothelial cell apoptosis.


Subject(s)
Angiotensin II/metabolism , Apoptosis/physiology , MicroRNAs/metabolism , Scavenger Receptors, Class E/metabolism , Angiotensin II/pharmacology , Apoptosis/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , MicroRNAs/genetics , Reactive Oxygen Species/metabolism , Scavenger Receptors, Class E/genetics , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...