Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters

Publication year range
1.
Brain Behav Immun ; 119: 919-944, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38718909

ABSTRACT

Neuroinflammation and accumulation of Amyloid Beta (Aß) accompanied by deterioration of special memory are hallmarks of Alzheimer's disease (AD). Effective preventative and treatment options for AD are still needed. Microglia in AD brains are characterized by elevated levels of microRNA-17 (miR-17), which is accompanied by defective autophagy, Aß accumulation, and increased inflammatory cytokine production. However, the effect of targeting miR-17 on AD pathology and memory loss is not clear. To specifically inhibit miR-17 in microglia, we generated mannose-coated lipid nanoparticles (MLNPs) enclosing miR-17 antagomir (Anti-17 MLNPs), which are targeted to mannose receptors readily expressed on microglia. We used a 5XFAD mouse model (AD) that recapitulates many AD-related phenotypes observed in humans. Our results show that Anti-17 MLNPs, delivered to 5XFAD mice by intra-cisterna magna injection, specifically deliver Anti-17 to microglia. Anti-17 MLNPs downregulated miR-17 expression in microglia but not in neurons, astrocytes, and oligodendrocytes. Anti-17 MLNPs attenuated inflammation, improved autophagy, and reduced Aß burdens in the brains. Additionally, Anti-17 MLNPs reduced the deterioration in spatial memory and decreased anxiety-like behavior in 5XFAD mice. Therefore, targeting miR-17 using MLNPs is a viable strategy to prevent several AD pathologies. This selective targeting strategy delivers specific agents to microglia without the adverse off-target effects on other cell types. Additionally, this approach can be used to deliver other molecules to microglia and other immune cells in other organs.


Subject(s)
Alzheimer Disease , Brain , Disease Models, Animal , Mannose , Mice, Transgenic , MicroRNAs , Microglia , Nanoparticles , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , MicroRNAs/metabolism , Nanoparticles/administration & dosage , Mice , Microglia/metabolism , Microglia/drug effects , Mannose/pharmacology , Brain/metabolism , Brain/drug effects , Amyloid beta-Peptides/metabolism , Lipids , Male , Antagomirs/pharmacology , Antagomirs/administration & dosage
2.
Mol Ther ; 29(6): 2041-2052, 2021 06 02.
Article in English | MEDLINE | ID: mdl-33609732

ABSTRACT

Oligonucleotide therapies offer precision treatments for a variety of neurological diseases, including epilepsy, but their deployment is hampered by the blood-brain barrier (BBB). Previous studies showed that intracerebroventricular injection of an antisense oligonucleotide (antagomir) targeting microRNA-134 (Ant-134) reduced evoked and spontaneous seizures in animal models of epilepsy. In this study, we used assays of serum protein and tracer extravasation to determine that BBB disruption occurring after status epilepticus in mice was sufficient to permit passage of systemically injected Ant-134 into the brain parenchyma. Intraperitoneal and intravenous injection of Ant-134 reached the hippocampus and blocked seizure-induced upregulation of miR-134. A single intraperitoneal injection of Ant-134 at 2 h after status epilepticus in mice resulted in potent suppression of spontaneous recurrent seizures, reaching a 99.5% reduction during recordings at 3 months. The duration of spontaneous seizures, when they occurred, was also reduced in Ant-134-treated mice. In vivo knockdown of LIM kinase-1 (Limk-1) increased seizure frequency in Ant-134-treated mice, implicating de-repression of Limk-1 in the antagomir mechanism. These studies indicate that systemic delivery of Ant-134 reaches the brain and produces long-lasting seizure-suppressive effects after systemic injection in mice when timed with BBB disruption and may be a clinically viable approach for this and other disease-modifying microRNA therapies.


Subject(s)
Antagomirs/genetics , Blood-Brain Barrier/metabolism , Epilepsy/genetics , Epilepsy/therapy , Animals , Antagomirs/administration & dosage , Blood-Brain Barrier/pathology , Disease Management , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation , Gene Silencing , Gene Transfer Techniques , Genetic Predisposition to Disease , Genetic Therapy , Mice , MicroRNAs/genetics , RNA Interference , Treatment Outcome
3.
Gastroenterology ; 159(3): 999-1014.e9, 2020 09.
Article in English | MEDLINE | ID: mdl-32450149

ABSTRACT

BACKGROUND & AIMS: Development of nonalcoholic steatohepatitis (NASH) is associated with reductions in hepatic microRNA122 (MIR122); the RAR related orphan receptor A (RORA) promotes expression of MIR122. Increasing expression of RORA in livers of mice increases expression of MIR122 and reduces lipotoxicity. We investigated the effects of a RORA agonist in mouse models of NASH. METHODS: We screened a chemical library to identify agonists of RORA and tested their effects on a human hepatocellular carcinoma cell line (Huh7). C57BL/6 mice were fed a chow or high-fat diet (HFD) for 4 weeks to induce fatty liver. Mice were given hydrodynamic tail vein injections of a MIR122 antagonist (antagomiR-122) or a control antagomiR once each week for 3 weeks while still on the HFD or chow diet, or intraperitoneal injections of the RORA agonist RS-2982 or vehicle, twice each week for 3 weeks. Livers, gonad white adipose, and skeletal muscle were collected and analyzed by reverse-transcription polymerase chain reaction, histology, and immunohistochemistry. A separate group of mice were fed an atherogenic diet, with or without injections of RS-2982 for 3 weeks; livers were analyzed by immunohistochemistry, and plasma was analyzed for levels of aminotransferases. We analyzed data from liver tissues from patients with NASH included in the RNA-sequencing databases GSE33814 and GSE89632. RESULTS: Injection of mice with antagomiR-122 significantly reduced levels of MIR122 in plasma, liver, and white adipose tissue; in mice on an HFD, antagomiR-122 injections increased fat droplets and total triglyceride content in liver and reduced ß-oxidation and energy expenditure, resulting in significantly more weight gain than in mice given the control microRNA. We identified RS-2982 as an agonist of RORA and found it to increase expression of MIR122 promoter activity in Huh7 cells. In mice fed an HFD or atherogenic diet, injections of RS-2982 increased hepatic levels of MIR122 precursors and reduced hepatic synthesis of triglycerides by reducing expression of biosynthesis enzymes. In these mice, RS-2982 significantly reduced hepatic lipotoxicity, reduced liver fibrosis, increased insulin resistance, and reduced body weight compared with mice injected with vehicle. Patients who underwent cardiovascular surgery had increased levels of plasma MIR122 compared to its levels before surgery; increased expression of plasma MIR122 was associated with increased levels of plasma free fatty acids and levels of RORA. CONCLUSIONS: We identified the compound RS-2982 as an agonist of RORA that increases expression of MIR122 in cell lines and livers of mice. Mice fed an HFD or atherogenic diet given injections of RS-2982 had reduced hepatic lipotoxicity, liver fibrosis, and body weight compared with mice given the vehicle. Agonists of RORA might be developed for treatment of NASH.


Subject(s)
Lipid Regulating Agents/pharmacology , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/drug therapy , Nuclear Receptor Subfamily 1, Group F, Member 1/agonists , Obesity/drug therapy , Animals , Antagomirs/administration & dosage , Benzamides/pharmacology , Benzamides/therapeutic use , Body Weight , Cell Line, Tumor , Datasets as Topic , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/metabolism , Humans , Insulin Resistance , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Lipid Regulating Agents/therapeutic use , Liver/drug effects , Liver/pathology , Male , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/blood , Mutation , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Promoter Regions, Genetic/drug effects , Up-Regulation/drug effects
4.
FASEB J ; 34(5): 6934-6949, 2020 05.
Article in English | MEDLINE | ID: mdl-32239566

ABSTRACT

Neutrophil infiltration and phenotypic transformation are believed to contribute to neuronal damage in ischemic stroke. Emerging evidence suggests that histone deacetylase 2 (HDAC2) is an epigenetic regulator of inflammatory cells. Here, we aimed to investigate whether microRNA-494 (miR-494) affects HDAC2-mediated neutrophil infiltration and phenotypic shift. MiR-494 levels in neutrophils from acute ischemic stroke (AIS) patients were detected by real-time PCR. Chromatin Immunoprecipitation (ChIP)-Seq was performed to clarify which genes are the binding targets of HDAC2. Endothelial cells and cortical neurons were subjected to oxygen-glucose deprivation (OGD), transwell assay was conducted to examine neutrophil migration through endothelial cells, and neuronal injury was examined after stimulating with supernatant from antagomiR-494-treated neutrophils. C57BL/6J mice were subjected to transient middle cerebral artery occlusion (MCAO) and antagomiR-494 was injected through tail vein immediately after reperfusion, and neutrophil infiltration and phenotypic shift was examined. We found that the expression of miR-494 in neutrophils was significantly increased in AIS patients. HDAC2 targeted multiple matrix metalloproteinases (MMPs) and Fc-gamma receptor III (CD16) genes in neutrophils of AIS patients. Furthermore, antagomiR-494 repressed expression of multiple MMPs genes, including MMP7, MMP10, MMP13, and MMP16, which reduced the number of brain-infiltrating neutrophils by regulating HDAC2. AntagomiR-494 could also exert its neuroprotective role through inhibiting the shift of neutrophils toward pro-inflammatory N1 phenotype in vivo and in vitro. Taken together, miR-494 may serve as an alternative predictive biomarker of the outcome of AIS patients, and antagomiR-494 treatment decreases the expression of multiple MMPs and the infiltration of neutrophils and inhibits the shift of neutrophils into N1 phenotype partly by targeting HDAC2.


Subject(s)
Antagomirs/administration & dosage , Histone Deacetylase 2/metabolism , MicroRNAs/antagonists & inhibitors , Neutrophils/metabolism , Stroke/therapy , Administration, Intravenous , Animals , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/therapy , Case-Control Studies , Disease Models, Animal , HL-60 Cells , Histone Deacetylase Inhibitors/administration & dosage , Humans , Male , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Neutrophils/drug effects , Neutrophils/pathology , RNA Interference , Stroke/genetics , Stroke/metabolism
5.
J Mol Cell Cardiol ; 140: 56-67, 2020 03.
Article in English | MEDLINE | ID: mdl-32135167

ABSTRACT

AIMS: Doxorubicin (DOX) is a broad-spectrum anticancer drug with considerable cardiotoxicity. DOX can induce myocardial apoptosis by modulating multiple signalling pathways. A better understanding of the underlying mechanism of DOX's cardiotoxicity will improve its clinical application and help avoid heart failure in patients. METHODS AND RESULTS: Models of DOX cardiotoxicity in cultured cardiomyocytes and mice were used. Cell death was determined by TUNEL and caspase 3/7 activity assay. Quaking (QKI) expression was detected by immunoblotting; microRNA-31-5p and circular RNA (circRNA) levels were determined by qRT-PCR. Luciferase reporter assays were performed to validate the miR-31-5p target. We found that DOX treatment upregulated miR-31-5p expression both in cultured cardiomyocytes and in mouse heart tissue. Silencing of miR-31-5p significantly alleviated the myocardial apoptosis induced by DOX treatment both in vivo and in vitro. Further analysis indicated QKI as a direct target of miR-31-5p, which has been reported to influence circRNA expression in a series of cell types. We found that circPan3 was specifically downregulated in cardiomyocytes upon DOX treatment. We further confirmed that the downregulation of circPan3 was due to the silencing of QKI by miR-31-5p. CONCLUSIONS: Our data reveal links among miR-31-5p, QKI and circPan3 in the apoptotic programme of cardiomyocytes. MiR-31-5p acted as a negative regulator of circPan3 by directly suppressing QKI, which may be a potential therapeutic target and strategy for DOX-induced cardiotoxicity.


Subject(s)
Cardiotoxicity/metabolism , Doxorubicin/pharmacology , MicroRNAs/metabolism , RNA, Circular/metabolism , RNA-Binding Proteins/metabolism , Animals , Antagomirs/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Carrier Proteins/genetics , Cell Line , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , RNA Interference , Rats , Transfection
6.
Dev Biol ; 445(2): 163-169, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30496744

ABSTRACT

BACKGROUND: Studying microRNA networks during heart development is essential to obtain a better understanding of developmental defects and diseases associated with the heart and to identify novel opportunities for therapeutics. Here we highlight the advantages of chicken embryos as a vertebrate model for studying intermediate processes of heart development. Avians develop a four-chambered heart closely resembling human anatomy and they develop ex utero, which makes them easily accessible. Furthermore, embryos are available all year with a steady supply. RESULTS: In this report we established a novel method for the knockdown of microRNA function by microinjecting AntagomiRs into the chicken heart in ovo. Our approach enables the targeted delivery of antagomirs into a locally restricted area and is not impacted by circulation. After further embryo development the successful miRNA knockdown was confirmed. Loss of function phenotypes can be evaluated rapidly, compared to more time-consuming genetic ablation experiments. The local application avoids potential systemic effects of microRNA knockdown, therefore allowing the assessment of impacts on heart development only. The method can be adjusted for different stages of chicken embryos (HH13-HH18) as well as for knockdown or targeted overexpression of coding genes. CONCLUSION: In conclusion our method allows targeted and locally restricted delivery of Antagomirs to the heart leading to successful knockdown of microRNA function. This method enables rapid phenotypic assessment, for example by gene expression analysis of multiple cardiac genes.


Subject(s)
Antagomirs/administration & dosage , Gene Knockdown Techniques/methods , Heart/embryology , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Animals , Chick Embryo , Heart Rate , Humans , Microinjections , Models, Animal , Organogenesis/genetics
7.
J Cell Physiol ; 235(10): 6887-6895, 2020 10.
Article in English | MEDLINE | ID: mdl-32003016

ABSTRACT

MicroRNAs are key factors for many biological functions. These regulatory molecules affect various gene networks and involve the subsequent signaling pathways. Therefore, disrupting the expression of these molecules is associated with multiple anomalies in the cells and body. One of the most important related abnormalities is the incidence of cancer. Thus, targeting microRNAs (miRNAs) is an effective approach for cancer gene therapy. Various factors are used for this purpose, including the antagomir nucleotide structure. There are some obstacles in the delivery of nucleotide therapeutics to the target cells, however, the use of nanoparticles could partly overcome these defeciencies. On the other hand, targeted delivery of antagomirs using aptamers, reduces nonspecific effects on nontarget cells. Considering the above, in this study, we designed and fabricated a nanocarrier composed of gold nanoparticles (GNPs), antagomir-155, and nucleolin specific aptamer for breast cancer study and therapy. Here, GNPs were synthesized using citrate reduction and were modified by polyA sequences, AS1411 aptamer, and antagomir-155. Attachment of molecules were confirmed using gel electrophoresis, atomic force microscopy imaging and electrochemical test. The specific entry of modified nanoparticles was investigated by fluorescence microscopy. The efficacy of modified nanoparticles was evaluated using a quantitative polymerase chain reaction (q-PCR) for miR-155 and its target gene. Efficient and specific delivery of AuNP-Apt-anti-miR-155 to target cells was confirmed in comparison with the control cell. The q-PCR analysis showed not only a significant decrease in mir-155 levels but also an elevated TP53INP1 mRNA, direct target of miR-155. The proposed structure inhibits proliferation and stimulates apoptosis by increasing the expression of TP53INP1. Our results suggest that AuNP-Apt-anti-miR-155 could be a promising nano constructor for breast cancer treatment.


Subject(s)
Antagomirs/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Gold/administration & dosage , Metal Nanoparticles/administration & dosage , MicroRNAs/antagonists & inhibitors , Oligodeoxyribonucleotides/administration & dosage , Animals , Antagomirs/chemistry , Apoptosis/drug effects , Aptamers, Nucleotide , CHO Cells , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cricetulus , Drug Delivery Systems/methods , Female , Gold/chemistry , Humans , MCF-7 Cells , Metal Nanoparticles/chemistry , Oligodeoxyribonucleotides/chemistry
8.
Respir Res ; 21(1): 186, 2020 Jul 16.
Article in English | MEDLINE | ID: mdl-32678044

ABSTRACT

BACKGROUND: The pathogenesis of pulmonary arterial hypertension (PAH) involves many signalling pathways. MicroRNAs are potential candidates involved in simultaneously coordinating multiple genes under such multifactorial conditions. METHODS AND RESULTS: MiR-138-5p is overexpressed in pulmonary arterial smooth muscle cells (PASMCs) from PAH patients and in lungs from rats with monocrotaline-induced pulmonary hypertension (MCT-PH). MiR-138-5p is predicted to regulate the expression of the potassium channel KCNK3, whose loss is associated with the development and progression of PAH. We hypothesized that, in vivo, miR-138-5p inhibition would restore KCNK3 lung expression and subsequently alleviate PAH. Nebulization-based delivery of anti-miR-138-5p to rats with established MCT-PH significantly reduced the right ventricular systolic pressure and significantly improved the pulmonary arterial acceleration time (PAAT). These haemodynamic improvements were related to decrease pulmonary vascular remodelling, lung inflammation and pulmonary vascular cell proliferation in situ. In vivo inhibition of miR-138-5p restored KCNK3 mRNA expression and SLC45A3 protein expression in the lungs. CONCLUSIONS: We confirmed that in vivo inhibition of miR-138-5p reduces the development of PH in experimental MCT-PH. The possible curative mechanisms involve at least the normalization of lung KCNK3 as well as SLC45A3 expression.


Subject(s)
Antagomirs/administration & dosage , Arterial Pressure , MicroRNAs/antagonists & inhibitors , Monosaccharide Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Pulmonary Arterial Hypertension/prevention & control , Pulmonary Artery/metabolism , Administration, Inhalation , Animals , Antagomirs/genetics , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Humans , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Monocrotaline , Monosaccharide Transport Proteins/genetics , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Artery/physiopathology , Rats, Wistar , Signal Transduction , Vascular Remodeling
9.
Cereb Cortex ; 29(4): 1509-1519, 2019 04 01.
Article in English | MEDLINE | ID: mdl-29522177

ABSTRACT

ß-Catenin has been implicated in major depressive disorder (MDD), which is associated with synaptic plasticity and dendritic arborization. MicroRNAs (miRNA) are small noncoding RNAs containing about 22 nucleotides and involved in a variety of physiological and pathophysiological process, but their roles in MDD remain largely unknown. Here, we investigated the expression and function of miRNAs in the mouse model of chronic social defeat stress (CSDS). The regulation of ß-catenin by selected miRNA was validated by silico prediction, target gene luciferase reporter assay, and transfection experiment in neurons. We demonstrated that the levels of miR-214-3p, which targets ß-catenin transcripts were significantly increased in the medial prefrontal cortex (mPFC) of CSDS mice. Antagomir-214-3p, a neutralizing inhibitor of miR-214-3p, increased the levels of ß-catenin and reversed the depressive-like behavior in CSDS mice. Meanwhile, antagomir-214-3p increased the amplitude of miniature excitatory postsynaptic current (mEPSC) and the number of dendritic spines in mPFC of CSDS mice, which may be related to the elevated expression of cldn1. Furthermore, intranasal administered antagomir-214-3p also significantly increased the level of ß-catenin and reversed the depressive-like behaviors in CSDS mice. These results may represent a new therapeutic target for MDD.


Subject(s)
Depression/physiopathology , MicroRNAs/physiology , Stress, Psychological/physiopathology , beta Catenin/physiology , Administration, Intranasal , Animals , Antagomirs/administration & dosage , Claudin-1/genetics , Dendritic Spines/drug effects , Dendritic Spines/physiology , Depression/etiology , Depression/genetics , Excitatory Postsynaptic Potentials/drug effects , Gene Expression Regulation , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Stress, Psychological/genetics , beta Catenin/genetics
10.
Med Sci Monit ; 26: e920394, 2020 Mar 14.
Article in English | MEDLINE | ID: mdl-32170053

ABSTRACT

BACKGROUND Oxidative stress and myocardial apoptosis are features of doxorubicin-induced cardiac toxicity that can result in cardiac dysfunction. Previous studies showed that microRNA-143 (miR-143) was expressed in the myocardium and had a role in cardiac function. This study aimed to investigate the effects and possible molecular mechanisms of miR-143 on oxidative stress and myocardial cell apoptosis in a mouse model of doxorubicin-induced cardiac toxicity. MATERIAL AND METHODS Mice underwent intraperitoneal injection of doxorubicin (15 mg/kg) daily for eight days to develop the mouse model of doxorubicin-induced cardiac toxicity. Four days before doxorubicin administration, a group of mice was pretreated daily with a miR-143 antagonist (25 mg/kg/day) for four consecutive days by tail vein injection. The study included the use of a miR-143 antagomir, or anti-microRNA, an oligonucleotide that silenced endogenous microRNA (miR), and an agomir to miR-143, and also the AKT inhibitor, MK2206. Quantitative real-time polymerase chain reaction (qRT-PCR) and immunoblot analysis were used to measure mRNA and protein expression, respectively. RESULTS Doxorubicin treatment increased the expression of miR-143, which was reduced by the miR-143 antagomir. Overexpression of miR-143 increased doxorubicin-induced myocardial apoptosis and oxidative stress. The use of the miR-143 antagomir significantly activated protein kinase B (PKB) and AKT, which were reduced in the presence of the AKT inhibitor, MK2206. However, the use of the miR-143 antagomir further down-regulated AKT phosphorylation following doxorubicin treatment and increased AKT activation. CONCLUSIONS In a mouse model of doxorubicin-induced cardiac toxicity, miR-143 increased oxidative stress and myocardial cell apoptosis following doxorubicin treatment by inhibiting AKT.


Subject(s)
Cardiotoxicity/genetics , Doxorubicin/toxicity , MicroRNAs/metabolism , Oxidative Stress/genetics , Animals , Antagomirs/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Cardiotoxicity/etiology , Cell Line , Disease Models, Animal , Heart/drug effects , Heterocyclic Compounds, 3-Ring/administration & dosage , Male , Mice , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects
11.
Med Sci Monit ; 26: e915825, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-32061171

ABSTRACT

BACKGROUND Ischemia-reperfusion injury is caused by a blood reperfusion injury in ischemic brain tissue, and usually occurs in the treatment stage of ischemic disease, which can aggravate brain tissue injury. MiR-122 is closely related to ischemia-reperfusion injury in the myocardium, kidney, and liver; however, the role in cerebral ischemia-reperfusion injury has not been established. MATERIAL AND METHODS In this study, cerebral ischemia-reperfusion injury was established in a rat model, and the control group was a sham-operated group. After ischemia-reperfusion injury for 6, 12, and 24 hours, brain tissue specimens were collected and the expression of miR-122 and DJ-1 were determined using quantitative real-time polymerase chain reaction. Flow cytometry was used to determine the reactive oxygen species (ROS) content. The modified Neurological Severity Score (mNSS) scale was used to evaluate the sensory and motor function defects of the rats. The malondialdehyde (MDA), superoxide dismutase (SOD), and enzyme activity were determined. The rats in the cerebral ischemia-reperfusion injury model were divided into 2 groups (antagomir-NC group and antagomir miR-122 group). Brain neuron RN-c cells were divided into the following 4 groups: antagomir-NC, antagomir miR-122, pIRES2-blank, and pIRES2-DJ-1. Seventy-two hours after transfection, ischemia-reperfusion treatment was carried out and conventional cultured RN-c cells were used as the control group. Flow cytometry was used to detect apoptosis and western blot was used to detect the expression of DJ-1, PTEN, AKT, and p-AKT. RESULTS The expression of miR-122 increased significantly in the process of ischemia-reperfusion damage after cerebral infarction, while the expression of DJ-1 decreased significantly. Downregulation of miR-122 significantly increased the expression of DJ-1, enhanced the activity of the PTEN/PI3K/AKT pathway, reduced cell apoptosis, and alleviated cerebral ischemia-reperfusion injury. CONCLUSIONS Inhibition of miR-122 can decrease cerebral ischemia-reperfusion injury by upregulating DJ-1-PTEN/PI3K/AKT pathway.


Subject(s)
Brain Ischemia/complications , MicroRNAs/metabolism , Protein Deglycase DJ-1/genetics , Reperfusion Injury/genetics , Signal Transduction/genetics , Animals , Antagomirs/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Brain/blood supply , Brain/drug effects , Brain/pathology , Brain Ischemia/drug therapy , Brain Ischemia/genetics , Brain Ischemia/pathology , Computational Biology , Disease Models, Animal , Humans , Male , MicroRNAs/antagonists & inhibitors , Oxidative Stress/drug effects , Oxidative Stress/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Reactive Oxygen Species/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Up-Regulation/drug effects
12.
J Stroke Cerebrovasc Dis ; 29(8): 104850, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32689640

ABSTRACT

BACKGROUND: Following the recent discovery that microRNA-134-5p (miR-134-5p) is elevated in the early stages of acute myocardial infarction (AMI), we examined the specific role of miR-134-5p in cardiomyocytes during AMI. METHODS: To study miR-134-5p's role in the context of AMI, we used a combination of in vitro experiments in H2O2-treated or hypoxic cardiomyocyte cell cultures as well as in vivo experiments in a murine model of AMI. RESULTS: H2O2- and hypoxia-induced cardiomyocyte injury upregulated miR-134-5p expression. miR-134-5p overexpression increased cardiomyocyte apoptosis, whereas miR-134-5p inhibition reduced cardiomyocyte apoptosis. We discovered that the transcription factor cAMP-responsive element binding protein 1 (Creb1) is a functional target of miR-134-5p responsible for regulating cardiomyocyte apoptosis. In vivo AMI resulted in the upregulation and downregulation of miR-134-5p and Creb1 in the infarct area, respectively. Circulating miR-134-5p levels were also increased at days 1 and 2 post-AMI. Modulation of myocardial miR-124-5p expression by intramyocardial injection of antagomiR-134-5p or agomiR-134-5p significantly affected cardiomyocyte apoptosis, infarct size, and cardiac function in vivo. CONCLUSIONS: miR-134-5p/Creb1 axis dysregulation plays a role in hypoxia- or oxidative stress-induced cardiomyocyte apoptosis as well as AMI. Circulating miR-134-5p may show promise as a biomarker for AMI or post-AMI cardiac dysfunction. Manipulating the miR-134-5p/Creb1 axis through either inhibition of miR-134-5p or overexpression of Creb1 may show promise as a novel therapeutic strategy to attenuate cardiac dysfunction following AMI.


Subject(s)
Antagomirs/administration & dosage , Apoptosis , Cyclic AMP Response Element-Binding Protein/metabolism , MicroRNAs/antagonists & inhibitors , Myocardial Infarction/prevention & control , Myocytes, Cardiac/metabolism , 3' Untranslated Regions , Animals , Binding Sites , Cell Hypoxia , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Hydrogen Peroxide/toxicity , JNK Mitogen-Activated Protein Kinases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Oxidative Stress , Signal Transduction , Up-Regulation
13.
Am J Physiol Heart Circ Physiol ; 317(3): H531-H540, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31225989

ABSTRACT

As an inflammatory disease afflicting the heart muscle, autoimmune myocarditis (AM) represents one of the foremost causes of heart failure. Accumulating evidence has implicated microRNAs (miRNAs) in the process of inflammation and autoimmunity. Hence, the current study aimed to investigate the mechanism by which miR-141-3p influences experimental AM (EAM). An EAM mouse model was established using 6-wk old male BALB/c mice, after which the expression of miR-141-3p and STAT4 was measured. Gain-of-function and loss-of-function investigations were performed to identify the functional role of miR-141-3p and STAT4 in EAM. Heart weight-to-body weight ratio, cardiac function, and degree of inflammation, as well as the levels of inflammation factors (IFN-γ, TNF-α, IL-2, IL-6, and IL-17) in the serum were detected. STAT4 was subsequently verified to be upregulated, and miR-141-3p was downregulated in the EAM mice. Furthermore, the overexpression of miR-141-3p or silencing of STAT4 was observed to reduce the heart weight-to-body weight ratio of EAM mice and improve cardiac function, while alleviating the degree of inflammatory cell infiltration in the myocardial tissue. Meanwhile, the overexpression of miR-141-3p was identified to diminish serum inflammatory factor levels by downregulating STAT4. Additionally, miR-141-3p could bind to STAT4 to downregulate its expression, ultimately mitigating inflammation and inducing an anti-inflammatory effect in EAM mice. Taken together, upregulation of miR-141-3p alleviates the inflammatory response in EAM mice by inhibiting STAT4, providing a promising intervention target for the molecular treatment of AM.NEW & NOTEWORTHY miR-141-3p is poorly expressed, and STAT4 is upregulated in experimental autoimmune myocarditis (EAM) mice. Overexpressing miR-141-3p inhibits EAM. miR-141-3p binds to and suppresses STAT4 expression. miR-141-3p overexpression inhibits inflammatory factors by downregulating STAT4. This study provides new insights into the treatment of autoimmune myocarditis.


Subject(s)
Autoimmune Diseases/prevention & control , MicroRNAs/metabolism , Myocarditis/prevention & control , Myocardium/metabolism , RNA, Small Interfering/administration & dosage , RNAi Therapeutics , STAT4 Transcription Factor/metabolism , Animals , Antagomirs/administration & dosage , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Cytokines/blood , Cytokines/genetics , Disease Models, Animal , Gene Regulatory Networks , Inflammation Mediators/blood , Male , Mice, Inbred BALB C , MicroRNAs/genetics , Myocarditis/genetics , Myocarditis/immunology , Myocarditis/metabolism , Myocardium/immunology , Myosins , RNA Interference , STAT4 Transcription Factor/genetics
14.
Pharm Res ; 36(10): 145, 2019 Aug 08.
Article in English | MEDLINE | ID: mdl-31396764

ABSTRACT

PURPOSE: The immediate plasma metabolism and development of chemo-resistance (single agent) severely hampers the clinical effectiveness of Sorafenib (SRF) in liver cancer therapy. MicroRNA27a inhibition is a promising biological strategy for breast cancer therapy. METHODS: In this study, we aimed to prepare SRF and anti-miRNA27a-loaded anti-GPC3 antibody targeted lipid nanoparticles to enhance the therapeutic efficacy against liver cancers. In this study, we have employed a unique cationic switchable lipid (CSL) as a mean to encapsulate miRNA as well as to confer pH-responsiveness to the nanocarrier system. RESULTS: The G-S27LN was nanosized and offered a pH-responsive release of SRF from the carrier system and we have demonstrated the specific affinity of G-S27LN towards the GPC3-overexpressed HepG2 cancer cells. Anti-microRNA27a significantly increased the protein expression of FOXO1 and PPAR-γ which are crucial components involved in proliferation and apoptosis of tumor cells. Combination of SRF and anti-miRNA27a (G-S27LN) resulted in significantly lower cell viability with a marked increase in the apoptosis cell proportion compared to that of free SRF indicating the synergistic anticancer effect. Animal studies in liver cancer xenograft model demonstrated significant suppression of tumor burden, reduced tumor cell and elevated TUNEL positive apoptosis with no toxicity concerns in animals treated with G-S27LN formulation. CONCLUSION: The CSL-based G-S27LN efficiently co-delivered anti-microRNA27a and SRF and therefore represents a promising therapy to treat liver cancer. This study also brings forth a platform strategy for the effective treatment of number of other advanced cancers.


Subject(s)
Antagomirs/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Glypicans/immunology , Lipids/chemistry , Liver Neoplasms/drug therapy , MicroRNAs/immunology , Nanoparticles/chemistry , Sorafenib/administration & dosage , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cholesterol/chemistry , Drug Synergism , Forkhead Box Protein O1/metabolism , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Mice, Inbred BALB C , Mice, Nude , PPAR gamma/metabolism , Phosphorylcholine/chemistry , Polyethylene Glycols/chemistry
15.
Mol Ther ; 26(8): 2008-2018, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29929788

ABSTRACT

We previously demonstrated that miR-214 is upregulated in malignant melanomas and triple-negative breast tumors and promotes metastatic dissemination by affecting a complex pathway including the anti-metastatic miR-148b. Importantly, tumor dissemination could be reduced by blocking miR-214 function or increasing miR-148b expression or by simultaneous interventions. Based on this evidence, with the intent to explore the role of miR-214 as a target for therapy, we evaluated the capability of new chemically modified anti-miR-214, R97/R98, to inhibit miR-214 coordinated metastatic traits. Relevantly, when melanoma or breast cancer cells were transfected with R97/R98, anti-miR-214 reduced miR-214 expression and impaired transendothelial migration were observed. Noteworthy, when the same cells were injected in the tail vein of mice, cell extravasation and metastatic nodule formation in lungs were strongly reduced. Thus, suggesting that R97/R98 anti-miR-214 oligonucleotides were able to inhibit tumor cell escaping through the endothelium. More importantly, when R97/R98 anti-miR-214 compounds were systemically delivered to mice carrying melanomas or breast or neuroendocrine pancreatic cancers, a reduced number of circulating tumor cells and lung or lymph node metastasis formation were detected. Similar results were also obtained when AAV8-miR-214 sponges were used in neuroendocrine pancreatic tumors. Based on this evidence, we propose miR-214 as a promising target for anti-metastatic therapies.


Subject(s)
Antagomirs/administration & dosage , MicroRNAs/genetics , Neoplasms/drug therapy , Up-Regulation/drug effects , Animals , Antagomirs/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/genetics , Cell Line, Tumor , Disease Progression , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Melanoma/drug therapy , Melanoma/genetics , Mice , MicroRNAs/antagonists & inhibitors , Neoplasm Metastasis/drug therapy , Neoplasms/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Transfection , Xenograft Model Antitumor Assays
16.
Mol Ther ; 26(9): 2178-2188, 2018 09 05.
Article in English | MEDLINE | ID: mdl-29802017

ABSTRACT

Lyophilized keratinocyte-targeted nanocarriers (TLNκ) loaded with locked nucleic acid (LNA) modified anti-miR were developed for topical application to full thickness burn injury. TLNκ were designed to selectively deliver LNA-anti-miR-107 to keratinocytes using the peptide sequence ASKAIQVFLLAG. TLNκ employed DOTAP/DODAP combination pH-responsive lipid components to improve endosomal escape. To minimize interference of clearance by non-targeted cells, especially immune cells in the acute wound microenvironment, surface charge was neutralized. Lyophilization was performed to extend the shelf life of the lipid nanoparticles (LNPs). Encapsulation efficiency of anti-miR in lyophilized TLNκ was estimated to be 96.54%. Cargo stability of lyophilized TLNκ was tested. After 9 days of loading with anti-miR-210, TLNκ was effective in lowering abundance of the hypoxamiR miR-210 in keratinocytes challenged with hypoxia. Keratinocyte uptake of DiD-labeled TLNκ was selective and exceeded 90% within 4 hr. Topical application of hydrogel-dispersed lyophilized TLNκ encapsulating LNA anti-miR-107 twice a week significantly accelerated wound closure and restoration of skin barrier function. TLNκ/anti-miR-107 application depleted miR-107 and upregulated dicer expression, which accelerated differentiation of keratinocytes. Expression of junctional proteins such as claudin-1, loricrin, filaggrin, ZO-1, and ZO-2 were significantly upregulated following TLNκ/anti-miR-107 treatment. These LNPs are promising as topical therapeutic agents in the management of burn injury.


Subject(s)
Burns/drug therapy , Freeze Drying , Lipids/chemistry , Nanoparticles/chemistry , Skin/pathology , Animals , Antagomirs/administration & dosage , Antagomirs/therapeutic use , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Filaggrin Proteins , Flow Cytometry , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Male , Mice , MicroRNAs/metabolism , Skin/drug effects , Wound Healing
17.
Metab Brain Dis ; 34(5): 1355-1363, 2019 10.
Article in English | MEDLINE | ID: mdl-31134481

ABSTRACT

Accumulation of extracellular amyloid-ß (Aß) in hippocampal subregions is a hallmark of Alzheimer's disease (AD), which promotes neuronal apoptosis, potentiates cognitive decline and play a causative role in AD pathogenesis. However, whether this process is controlled by distinct miRNAs at the posttranscriptional level remain fascinating but poorly understood. Using post mortem hippocampal samples from human AD patients and 3xTg-AD mouse, we demonstrate that miR-342-3p expression was significantly induced during the AD development. With the aid of intrahippocampal injection of miR-342-3p antagomir, we further show that in vivo miR-342-3p inhibition synergistically improved cognitive deficits in 3xTg-AD mice. The hippocampal Aß-plaque burden in 3xTg-AD mice, as revealed by immunohistochemical analysis with 4G8 antibody, was attenuated also. Mechanistically, the upregulation of neuronal miR-342-3p is linked to an increase in the activation of the stress kinase c-Jun N-terminal kinase with the subsequent death of the neurons in Aß-challenged HT22 hippocampal neuronal cells. These findings support the model that derangement of hippocampus signal transduction and subsequent neuronal apoptosis in AD arises as a consequence of increased Aß burden and chronic activation of the JNK MAPK cascade in a miR-342-3p-dependent manner. Overall, we described for the first time the regulatory activity of miR-342-3p on relevant Aß metabolism pathways in Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Antagomirs/therapeutic use , Hippocampus/drug effects , Learning/drug effects , Memory/drug effects , MicroRNAs/antagonists & inhibitors , Plaque, Amyloid/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Antagomirs/administration & dosage , Cell Line , Female , Hippocampus/metabolism , Humans , Male , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
18.
Int J Mol Sci ; 20(16)2019 Aug 13.
Article in English | MEDLINE | ID: mdl-31412612

ABSTRACT

INTRODUCTION: MiRNAs have been shown to play a crucial role among lung cancer, pulmonary fibrosis, tuberculosis (TBC) infection, and bronchial hypersensitivity, thus including chronic obstructive pulmonary disease (COPD) and asthma. The oncogenic effect of several miRNAs has been recently ruled out. In order to act on miRNAs turnover, antagomiRs have been developed. MATERIALS AND METHODS: The systematic review was conducted under the PRISMA guidelines (registration number is: CRD42019134173). The PubMed database was searched between 1 January 2000 and 30 April 2019 under the following search strategy: (((antagomiR) OR (mirna antagonists) OR (mirna antagonist)) AND ((lung[MeSH Terms]) OR ("lung diseases"[MeSH Terms]))). We included original articles, published in English, whereas exclusion criteria included reviews, meta-analyses, single case reports, and studies published in a language other than English. RESULTS AND CONCLUSIONS: A total of 68 articles matching the inclusion criteria were retrieved. Overall, the use of antagomiR was seen to be efficient in downregulating the specific miRNA they are conceived for. The usefulness of antagomiRs was demonstrated in humans, animal models, and cell lines. To our best knowledge, this is the first article to encompass evidence regarding miRNAs and their respective antagomiRs in the lung, in order to provide readers a comprehensive review upon major lung disorders.


Subject(s)
Antagomirs/genetics , Gene Expression Regulation , Lung Diseases/genetics , RNA Interference , Animals , Antagomirs/administration & dosage , Biomarkers , Cell Line , Cells, Cultured , Humans , Lung Diseases/diagnosis , Lung Diseases/metabolism , Lung Diseases/therapy , MicroRNAs/genetics , Models, Animal
19.
Glia ; 66(7): 1346-1362, 2018 07.
Article in English | MEDLINE | ID: mdl-29451327

ABSTRACT

Reactive astrocytes induced by ischemia can transdifferentiate into mature neurons. This neurogenic potential of astrocytes may have therapeutic value for brain injury. Epigenetic modifications are widely known to involve in developmental and adult neurogenesis. PAX6, a neurogenic fate determinant, contributes to the astrocyte-to-neuron conversion. However, it is unclear whether microRNAs (miRs) modulate PAX6-mediated astrocyte-to-neuron conversion. In the present study we used bioinformatic approaches to predict miRs potentially targeting Pax6, and transient middle cerebral artery occlusion (MCAO) to model cerebral ischemic injury in adult rats. These rats were given striatal injection of glial fibrillary acidic protein targeted enhanced green fluorescence protein lentiviral vectors (Lv-GFAP-EGFP) to permit cell fate mapping for tracing astrocytes-derived neurons. We verified that miR-365 directly targets to the 3'-UTR of Pax6 by luciferase assay. We found that miR-365 expression was significantly increased in the ischemic brain. Intraventricular injection of miR-365 antagomir effectively increased astrocytic PAX6 expression and the number of new mature neurons derived from astrocytes in the ischemic striatum, and reduced neurological deficits as well as cerebral infarct volume. Conversely, miR-365 agomir reduced PAX6 expression and neurogenesis, and worsened brain injury. Moreover, exogenous overexpression of PAX6 enhanced the astrocyte-to-neuron conversion and abolished the effects of miR-365. Our results demonstrate that increase of miR-365 in the ischemic brain inhibits astrocyte-to-neuron conversion by targeting Pax6, whereas knockdown of miR-365 enhances PAX6-mediated neurogenesis from astrocytes and attenuates neuronal injury in the brain after ischemic stroke. Our findings provide a foundation for developing novel therapeutic strategies for brain injury.


Subject(s)
Astrocytes/metabolism , MicroRNAs/metabolism , Neurogenesis/physiology , Neurons/metabolism , PAX6 Transcription Factor/metabolism , Stroke/metabolism , Animals , Antagomirs/administration & dosage , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Hypoxia/physiology , Cells, Cultured , Disease Models, Animal , Glucose/deficiency , Male , MicroRNAs/antagonists & inhibitors , Neurons/pathology , Rats, Sprague-Dawley , Stroke/pathology
20.
Am J Physiol Gastrointest Liver Physiol ; 314(1): G109-G118, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29051187

ABSTRACT

In these studies, we developed a novel approach of in vivo magnetofection for localized delivery of nucleic acids such as micro-RNA-139-5p (miR-139-5p; which is known to target Rho kinase2) to the circular smooth muscle layer of the internal anal sphincter (IAS). The IAS tone is known to play a major role in the rectoanal continence via activation of RhoA-associated kinase (RhoA/ROCK2). These studies established an optimized protocol for efficient gene delivery using an assembly of equal volumes of in vivo PolyMag and miR139-5p or anti-miR-139-5p (100 nM each) injected in the circular smooth muscle layer in the pinpointed areas of the rat perianal region and then incubated for 20 min under magnetic field. Magnetofection efficiency was confirmed and analyzed by confocal microscopy of FITC-tagged siRNA. Using physiological and biochemical approaches, we investigated the effects of miR-139-5p and anti-miR-139-5p on basal intraluminal IAS pressure (IASP), fecal pellet count, IAS tone, agonist-induced contraction, contraction-relaxation kinetics, and RhoA/ROCK2 signaling. Present studies demonstrate that magnetofection-mediated miR-139-5p delivery significantly decreased RhoA/ROCK2, p-MYPT1, and p-MLC20 signaling, leading to decreases in the basal IASP and IAS tone and in rates of contraction and relaxation associated with increase in fecal pellet output. Interestingly, anti-miR-139-5p transfection had opposite effects on these parameters. Collectively, these data demonstrate that magnetofection is a promising novel method of in vivo gene delivery and of nucleotides to the internal anal sphincter for the site-directed and targeted therapy for rectoanal motility disorders. NEW & NOTEWORTHY These studies for the first time demonstrate the success of topical in vivo magnetofection (MF) of nucleic acids using perianal injections. To demonstrate its effectiveness, we used FITC-tagged siRNA via immunofluorescence microcopy and functional and biochemical evidence using miR-139-5p (which is known to target ROCK2). In conclusion, MF allows safe, convenient, efficient, and targeted delivery of oligonucleotides such as siRNAs and microRNAs. These studies have direct therapeutic implications in rectoanal motility disorders especially associated with IAS.


Subject(s)
Anal Canal/metabolism , Antagomirs/administration & dosage , Anus Diseases/therapy , Gastrointestinal Motility , Gene Transfer Techniques , Magnetics/methods , Magnetite Nanoparticles , MicroRNAs/administration & dosage , Animals , Antagomirs/genetics , Antagomirs/metabolism , Anus Diseases/genetics , Anus Diseases/metabolism , Anus Diseases/physiopathology , Defecation , Injections , Kinetics , MicroRNAs/genetics , MicroRNAs/metabolism , Myosin Light Chains/metabolism , Phosphorylation , Pressure , Protein Phosphatase 1/metabolism , Rats, Sprague-Dawley , Signal Transduction , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL