Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Arterioscler Thromb Vasc Biol ; 39(2): 250-262, 2019 02.
Article in English | MEDLINE | ID: mdl-30567484

ABSTRACT

Objective- TCF7L2 (transcription factor 7-like 2) is a Wnt-regulated transcription factor that maintains stemness and promotes proliferation in embryonic tissues and adult stem cells. Mice with a coronary artery disease-linked mutation in Wnt-coreceptor LRP6 (LDL receptor-related protein 6) exhibit vascular smooth muscle cell dedifferentiation and obstructive coronary artery disease, which are paradoxically associated with reduced TCF7L2 expression. We conducted a comprehensive study to explore the role of TCF7L2 in vascular smooth muscle cell differentiation and protection against intimal hyperplasia. Approach and Results- Using multiple mouse models, we demonstrate here that TCF7L2 promotes differentiation and inhibits proliferation of vascular smooth muscle cells. TCF7L2 accomplishes these effects by stabilization of GATA6 (GATA-binding protein 6) and upregulation of SM-MHC (smooth muscle cell myosin heavy chain) and cell cycle inhibitors. Accordingly, TCF7L2 haploinsufficient mice exhibited increased susceptibility to injury-induced hyperplasia, while mice overexpressing TCF7L2 were protected against injury-induced intimal hyperplasia compared with wild-type littermates. Consequently, the overexpression of TCF7L2 in LRP6 mutant mice rescued the injury-induced intimal hyperplasia. Conclusions- Our novel findings imply cell type-specific functional role of TCF7L2 and provide critical insight into mechanisms underlying the pathogenesis of intimal hyperplasia.


Subject(s)
Cell Plasticity , GATA6 Transcription Factor/physiology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Transcription Factor 7-Like 2 Protein/physiology , Tunica Intima/pathology , Animals , Cells, Cultured , Hyperplasia , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Platelet-Derived Growth Factor/pharmacology
2.
J Biol Chem ; 292(40): 16554-16570, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28842502

ABSTRACT

CAPA peptides, such as periviscerokinin (PVK), are insect neuropeptides involved in many signaling pathways controlling, for example, metabolism, behavior, and reproduction. They are present in a large number of insects and, together with their cognate receptors, are important for research into approaches for improving insect control. However, the CAPA receptors in the silkworm (Bombyx mori) insect model are unknown. Here, we cloned cDNAs of two putative CAPA peptide receptor genes, BNGR-A27 and -A25, from the brain of B. mori larvae. We found that the predicted BNGR-A27 ORF encodes 450 amino acids and that one BNGR-A25 splice variant encodes a full-length isoform (BNGR-A25L) of 418 amino acid residues and another a short isoform (BNGR-A25S) of 341 amino acids with a truncated C-terminal tail. Functional assays indicated that both BNGR-A25L and -A27 are activated by the PVK neuropeptides Bom-CAPA-PVK-1 and -PVK-2, leading to a significant increase in cAMP-response element-controlled luciferase activity and Ca2+ mobilization in a Gq inhibitor-sensitive manner. In contrast, BNGR-A25S was not significantly activated in response to the PVK peptides. Moreover, Bom-CAPA-PVK-1 directly bound to BNGR-A25L and -A27, but not BNGR-A25S. Of note, CAPA-PVK-mediated ERK1/2 phosphorylation and receptor internalization confirmed that BNGR-A25L and -A27 are two canonical receptors for Bombyx CAPA-PVKs. However, BNGR-A25S alone is a nonfunctional receptor but serves as a dominant-negative protein for BNGR-A25L. These results provide evidence that BNGR-A25L and -A27 are two functional Gq-coupled receptors for Bombyx CAPA-PVKs, enabling the further elucidation of the endocrinological roles of Bom-CAPA-PVKs and their receptors in insect biology.


Subject(s)
Bombyx , Calcium Signaling/physiology , Insect Proteins , Neuropeptides , Receptors, G-Protein-Coupled , Animals , Bombyx/genetics , Bombyx/metabolism , Insect Proteins/genetics , Insect Proteins/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
3.
Cell Physiol Biochem ; 45(2): 547-557, 2018.
Article in English | MEDLINE | ID: mdl-29402834

ABSTRACT

BACKGROUND/AIMS: Endothelial cell dysfunction is the principal pathological process underlying atherosclerotic cardiovascular disease. G protein-coupled receptor 124 (GPR124), an orphan receptor in the adhesion GPCR subfamily, promotes angiogenesis in the brain. In the present study, we explored the role of endothelial GPR124 in the development and progression of atherosclerosis in adult mice. METHODS: Using tetracycline-inducible transgenic systems, we generated mice expressing GPR124 specifically under control of the Tie-2 promoter. The animal model of atherosclerosis was constructed by intravenously injecting AAV-PCSK9DY into tetracycline-regulated mice and feeding the mice a high-fat diet for 16 consecutive weeks. Biochemical analysis and immunohistochemistry methods were used to address the role and mechanism of GPR124 in the pathological process of atherosclerosis. RESULTS: Higher TC (total cholesterol) and LDL-C (low density lipoprotein cholesterol) levels in serum and greater lipid deposition in the aortic sinus were found in atherosclerotic mice with GPR124 overexpression, coincident with the elevated proliferation of smooth muscle cells. We observed an elevation of ONOO- in the aortic sinus in this model by using immunofluorescence, and the experiments showed that the specific overexpression of GPR124 in the endothelium induced the up-regulation of CD68, NLRP3 and caspase-1 levels in the aortic sinus. CONCLUSION: The above results indicate that manipulating GPR124 in the endothelium may contribute to delayed pathological progression of atherosclerosis.


Subject(s)
Atherosclerosis/pathology , Receptors, G-Protein-Coupled/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Atherosclerosis/metabolism , Caspase 1/metabolism , Cholesterol/blood , Cholesterol, LDL/blood , Diet, High-Fat , Disease Models, Animal , Humans , Inflammation/etiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peroxynitrous Acid/metabolism , Plasmids/genetics , Plasmids/metabolism , Receptors, G-Protein-Coupled/genetics , Sinus of Valsalva/metabolism , Sinus of Valsalva/pathology
4.
J Am Chem Soc ; 137(38): 12296-303, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26352914

ABSTRACT

Accumulating evidence suggests that formation of peroxynitrite (ONOO(-)) in the cerebral vasculature contributes to the progression of ischemic damage, while the underlying molecular mechanisms remain elusive. To fully understand ONOO(-) biology, efficient tools that can realize the real-time tracing of endogenous ONOO(-) fluxes are indispensable. While a few ONOO(-) fluorescent probes have been reported, direct visualization of ONOO(-) fluxes in the cerebral vasculature of live mice remains a challenge. Herein, we present a fluorescent switch-on probe (NP3) for ONOO(-) imaging. NP3 exhibits good specificity, fast response, and high sensitivity toward ONOO(-) both in vitro and in vivo. Moreover, NP3 is two-photon excitable and readily blood-brain barrier penetrable. These desired photophysical and pharmacokinetic properties endow NP3 with the capability to monitor brain vascular ONOO(-) generation after injury with excellent temporal and spatial resolution. As a proof of concept, NP3 has enabled the direct visualization of neurovascular ONOO(-) formation in ischemia progression in live mouse brain by use of two-photon laser scanning microscopy. Due to these favorable properties, NP3 holds great promise for visualizing endogenous peroxynitrite fluxes in a variety of pathophysiological progressions in vitro and in vivo.


Subject(s)
Cerebrovascular Trauma/metabolism , Endothelial Cells/metabolism , Fluorescent Dyes/chemistry , Peroxynitrous Acid/metabolism , Animals , Cerebrovascular Trauma/pathology , Endothelial Cells/chemistry , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/pharmacokinetics , Mice , Molecular Structure , Peroxynitrous Acid/chemistry
5.
J Pineal Res ; 56(1): 1-11, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23952810

ABSTRACT

Lower global cognitive function scores are a common symptom of autism spectrum disorders (ASDs). This study investigates the effects of melatonin on hippocampal serine/threonine kinase signaling in an experimental ASD model. We found that chronic melatonin (1.0 or 5.0 mg/kg/day, 28 days) treatment significantly rescued valproic acid (VPA, 600 mg/kg)-induced decreases in CaMKII (Thr286), NMDAR1 (Ser896), and PKA (Thr197) phosphorylation in the hippocampus without affecting total protein levels. Compared with control rats, the immunostaining of pyramidal neurons in the hippocampus revealed a decrease in immunolabeling intensity for phospho-CaMKII (Thr286) in the hippocampus of VPA-treated rats, which was ameliorated by chronic melatonin treatment. Consistent with the elevation of CaMKII/PKA/PKC phosphorylation observed in melatonin-treated rat, long-term potentiation (LTP) was enhanced after chronic melatonin (5.0 mg/kg) treatment, as reflected by extracellular field potential slopes that increased from 56 to 60 min (133.4 ± 3.9% of the baseline, P < 0.01 versus VPA-treated rats) following high-frequency stimulation (HFS) in hippocampal slices. Accordingly, melatonin treatment also significantly improved social behavioral deficits at postnatal day 50 in VPA-treated rats. Taken together, the increased phosphorylation of CaMKII/PKA/PKC signaling might contribute to the beneficial effects of melatonin on autism symptoms.


Subject(s)
Autistic Disorder , Behavior, Animal/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/drug effects , Hippocampus/enzymology , Melatonin/pharmacology , Analysis of Variance , Animals , Antioxidants/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/analysis , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Disease Models, Animal , Female , Hippocampus/chemistry , Immunohistochemistry , Male , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Valproic Acid/pharmacology
6.
Nanomedicine ; 10(8): 1843-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24768629

ABSTRACT

The present study was designed to investigate the role of autophagy-lysosome signaling in the brain after application of nanoparticles. Here, lipid nanoparticles (LNs) induced elevations of Atg5, P62, LC3 and cathepsin B in mice brain. The transmission electron microscopy revealed a dramatic elevation of lysosome vacuoles colocalized with LNs cluster inside the neurons in mice brain. Immunoblot data revealed abnormal expression of cathepsin B in brain cortex following LNs injection, whereas its expression was further elevated in Atg5(+/-) mice. The importance of Atg5 in the LNs-induced autophagy-lysosome cascade was further supported by our finding that neurovascular response was exaggerated in Atg5(+/-) mice. In addition, the siRNA knockdown of Atg5 significantly blunted the increasing of LC3 and P62 in LNs-treated Neuro-2a cells. Taken together, we propose that LNs induce autophagy-lysosome signaling and neurovascular response at least partially via an Atg5-dependent pathway. FROM THE CLINICAL EDITOR: These authors investigated autophagy-lysosome signaling in the mouse brain after application of lipid nanoparticles and report that these nanoparticles induce autophagy-lysosome signaling and neurovascular response at least partially via an Atg5-dependent pathway.


Subject(s)
Brain/metabolism , Cathepsin B/metabolism , Lipids/chemistry , Lysosomes/metabolism , Microtubule-Associated Proteins/deficiency , Nanoparticles/chemistry , Animals , Autophagy-Related Protein 5 , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microtubule-Associated Proteins/genetics
7.
Sci China Life Sci ; 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39048715

ABSTRACT

CRISPR technologies have revolutionized research areas ranging from fundamental science to translational medicine. CRISPR-based genetic screens offer a powerful platform for unbiased screening in various fields, such as cancer immunology. Immune checkpoint blockade (ICB) therapy has been shown to strongly affect cancer treatment. However, the currently available ICBs are limited and do not work in all cancer patients. Pooled CRISPR screens enable the identification of previously unknown immune regulators that can regulate T-cell activation, cytotoxicity, persistence, infiltration into tumors, cytokine secretion, memory formation, T-cell metabolism, and CD4+ T-cell differentiation. These novel targets can be developed as new immunotherapies or used with the current ICBs as new combination therapies that may yield synergistic efficacy. Here, we review the progress made in the development of CRISPR technologies, particularly technological advances in CRISPR screens and their application in novel target identification for immunotherapy.

8.
Bioorg Med Chem ; 21(19): 5936-44, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23978359

ABSTRACT

A series of novel 3-substituted-indole derivatives with a benzyl tertiary amino moiety were designed, synthesized and evaluated as H3 receptor antagonists and free radical scavengers for Alzheimer's disease therapy. Most of these synthesized compounds exhibited moderate to potent antagonistic activities in CREs driven luciferase assay. In particular, compound 2d demonstrated the most favorable H3 receptor antagonistic activity with the IC50 value of 0.049µM. Besides, it also displayed high binding affinity to H3 receptor (Ki=4.26±2.55nM) and high selectivity over other three histamine receptors. Moreover, 2d and other two 3-substituted indole derivatives 1d and 3d exerted potent ABTS radical cation scavenging capacities similar to melatonin. Above results illustrate that 2d is an interesting lead for extensive optimization to explore new drug candidate for AD therapy.


Subject(s)
Drug Design , Free Radical Scavengers/chemical synthesis , Histamine Antagonists/chemical synthesis , Indoles/chemical synthesis , Receptors, Histamine H3 , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Histamine Antagonists/chemistry , Histamine Antagonists/pharmacology , Humans , Indoles/chemistry , Indoles/pharmacology , Inhibitory Concentration 50 , Models, Biological , Molecular Structure , Protein Binding/drug effects , Receptors, Histamine H3/chemistry , Sequence Homology , Structure-Activity Relationship
9.
J Clin Invest ; 133(4)2023 02 15.
Article in English | MEDLINE | ID: mdl-36602864

ABSTRACT

Genetic variants in the third intron of the PRDM6 gene have been associated with BP traits in multiple GWAS. By combining fine mapping, massively parallel reporter assays, and gene editing, we identified super enhancers that drive the expression of PRDM6 and are partly regulated by STAT1 as the causal variants for hypertension. The heterozygous disruption of Prdm6 in mice expressing Cre recombinase under the control of mouse smooth muscle cell protein 22-α promoter (Prdm6fl/+ SM22-Cre) exhibited a markedly higher number of renin-producing cells in the kidneys at E18.5 compared with WT littermates and developed salt-induced systemic hypertension that was completely responsive to the renin inhibitor aliskiren. Strikingly, RNA-Seq analysis of the mouse aortas identified a network of PRDM6-regulated genes that are located in GWAS-associated loci for blood pressure, most notably Sox6, which modulates renin expression in the kidney. Accordingly, the smooth muscle cell-specific disruption of Sox6 in Prdm6fl/+ SM22-Cre mice resulted in a dramatic reduction of renin. Fate mapping and histological studies also showed increased numbers of neural crest-derived cells accompanied by increased collagen deposition in the kidneys of Prdm6fl/+ Wnt1Cre-ZsGreen1Cre mice compared with WT mice. These findings establish the role of PRDM6 as a regulator of renin-producing cell differentiation into smooth muscle cells and as an attractive target for the development of antihypertensive drugs.


Subject(s)
Hypertension , Renin , Mice , Animals , Renin/genetics , Systems Biology , Hypertension/metabolism , Kidney/metabolism , Blood Pressure
10.
JCI Insight ; 7(4)2022 02 02.
Article in English | MEDLINE | ID: mdl-35108221

ABSTRACT

The molecular mechanisms that drive the acquisition of distinct neural crest cell (NCC) fates is still poorly understood. Here, we identified Prdm6 as an epigenetic modifier that temporally and spatially regulates the expression of NCC specifiers and determines the fate of a subset of migrating cardiac NCCs (CNCCs). Using transcriptomic analysis and genetic and fate mapping approaches in transgenic mice, we showed that disruption of Prdm6 was associated with impaired CNCC differentiation, delamination, and migration and led to patent ductus arteriosus (DA) and ventricular noncompaction. Bulk and single-cell RNA-Seq analyses of the DA and CNCCs identified Prdm6 as a regulator of a network of CNCC specification genes, including Wnt1, Tfap2b, and Sox9. Loss of Prdm6 in CNCCs diminished its expression in the pre-epithelial-mesenchymal transition (pre-EMT) cluster, resulting in the retention of NCCs in the dorsal neural tube. This defect was associated with diminished H4K20 monomethylation and G1-S progression and augmented Wnt1 transcript levels in pre-EMT and neural tube clusters, which we showed was the major driver of the impaired CNCC migration. Altogether, these findings revealed Prdm6 as a key regulator of CNCC differentiation and migration and identified Prdm6 and its regulated network as potential targets for the treatment of congenital heart diseases.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Developmental , Heart Defects, Congenital/genetics , Neural Crest/pathology , Organogenesis/genetics , RNA/genetics , Repressor Proteins/genetics , Animals , Cell Differentiation , Cell Movement , Disease Models, Animal , Female , Heart Defects, Congenital/metabolism , Mice , Mice, Knockout , Neural Crest/metabolism , Repressor Proteins/metabolism
11.
CNS Neurosci Ther ; 25(4): 476-485, 2019 04.
Article in English | MEDLINE | ID: mdl-30328295

ABSTRACT

AIMS: Autism spectrum disorder (ASD) is a wide range of neurodevelopmental disorders involving deficits in social interaction and communication. Unfortunately, autism remains a scientific and clinical challenge owing to the lack of understanding the cellular and molecular mechanisms underlying it. This study aimed to investigate the pathophysiological mechanism underlying leukocyte-endothelial adhesion in autism-related neurovascular inflammation. METHODS: Male BTBR T+tf/J mice were used as an autism model. The dynamic pattern of leukocyte-endothelial adhesion in mouse cerebral vessels was detected by two-photon laser scanning microscopy (TPLSM). Using FACS, RT-PCR, and Western blotting, we explored the expression of cell adhesion molecules, the mRNA expression of endothelial chemokine, the protein levels of cathepsin B, and inflammatory mediators. RESULTS: We found a significant increase in leukocyte-endothelial adhesion in BTBR mice, accompanied by elevated expression of the adhesion molecule neutrophils CD11b and endothelial ICAM-1. Our data further indicate that elevated neutrophil cathepsin B levels contribute to elevated endothelial chemokine CXCL7 levels in BTBR mice. The pharmacological inhibition of cathepsin B reverses the enhanced leukocyte-endothelial adhesion in the cerebral vessels of autistic mice. CONCLUSION: Our results revealed the prominent role of cathepsin B in modulating leukocyte-endothelial adhesion during autism-related neurovascular inflammation and identified a promising novel approach for autism treatment.


Subject(s)
Autistic Disorder/drug therapy , Cathepsin B/antagonists & inhibitors , Cell Adhesion/drug effects , Dipeptides/pharmacology , Endothelium, Vascular/drug effects , Leukocytes/drug effects , Animals , Autistic Disorder/metabolism , Cathepsin B/metabolism , Cell Adhesion/physiology , Dipeptides/therapeutic use , Disease Models, Animal , Endothelium, Vascular/metabolism , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
Theranostics ; 9(20): 5937-5955, 2019.
Article in English | MEDLINE | ID: mdl-31534530

ABSTRACT

Prolonged occlusion of multiple microvessels causes microvascular injury. G protein-coupled receptor 124 (GPR124) has been reported to be required for maintaining central nervous system (CNS) angiogenesis and blood-brain barrier integrity. However, the molecular mechanisms by which GPR124 regulates pericytes during ischemia have remained elusive. Methods: A microsphere embolism-induced ischemia model was used to evaluate the expression of GPR124 following microsphere embolism. Immunocytochemistry and stochastic optical reconstruction microscopy imaging were used to assess the expression and distribution of GPR124 in human brain vascular pericytes (HBVPs) and after the treatment with 3-morpholino-sydnonimine (SIN-1) or oxygen-glucose deprivation (OGD). The effect of GPR124 knockdown or overexpression on HBVP migration was analyzed in vitro using wound healing assays and a microfluidic device. GPR124 loss-of-function studies were performed in HBVPs and HEK293 cells using CRISPR-Cas9-mediated gene deletion. Time-lapse imaging was used to assess dynamic changes in the formation of filopodia in an individual cell. Finally, to explore the functional domains required for GPR124 activity, deletion mutants were constructed for each of the N-terminal domains. Results: GPR124 expression was increased in pericytes following microsphere embolism. Morphological analysis showed localization of GPR124 to focal adhesions where GPR124 bound directly to the actin binding protein vinculin and upregulated Cdc42. SIN-1 or OGD treatment redistributed GPR124 to the leading edges of HBVPs where GPR124 signaling was required for pericyte filopodia formation and directional migration. Partial deletion of GPR124 domains decreased SIN-1-induced filopodia formation and cell migration. Conclusion: Taken together, our results provide the first evidence for a role of GPR124 in pericyte migration under ischemic conditions and suggest that GPR124 was essential for Cdc42 activation and filopodia formation.


Subject(s)
Brain Ischemia/metabolism , Cell Polarity/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Pericytes/cytology , Pericytes/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Blotting, Western , Cell Line , Cell Polarity/genetics , Focal Adhesions/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Lentivirus/genetics , Male , Mice , Plasmids/genetics , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Wound Healing/genetics , Wound Healing/physiology
13.
Cell Discov ; 4: 46, 2018.
Article in English | MEDLINE | ID: mdl-30062046

ABSTRACT

CRISPR systems have been proven as versatile tools for site-specific genome engineering in mammalian species. During the gene editing processes, these RNA-guide nucleases introduce DNA double strand breaks (DSBs), in which non-homologous DNA end joining (NHEJ) dominates the DNA repair pathway, limiting the efficiency of homology-directed repair (HDR), the alternative pathway essential for precise gene targeting. Multiple approaches have been developed to enhance HDR, including chemical compound or RNA interference-mediated inhibition of NHEJ factors, small molecule activation of HDR enzymes, or cell cycle timed delivery of CRISPR complex. However, these approaches face multiple challenges, yet have moderate or variable effects. Here we developed a new approach that programs both NHEJ and HDR pathways with CRISPR activation and interference (CRISPRa/i) to achieve significantly enhanced HDR efficiency of CRISPR-mediated gene editing. The manipulation of NHEJ and HDR pathway components, such as CtIP, CDK1, KU70, KU80, and LIG4, was mediated by catalytically dead guide RNAs (dgRNAs), thus relying on only a single catalytically active Cas9 to perform both CRISPRa/i and precise gene editing. While reprogramming of most DNA repair factors or their combinations tested enhanced HDR efficiency, simultaneously activating CDK1 and repressing KU80 has the strongest effect with increased HDR rate upto an order of magnitude. Doxycycline-induced dgRNA-based CRISPRa/i programming of DNA repair enzymes, as well as viral packaging enabled flexible and tunable HDR enhancement for broader applicability in mammalian cells. Our study provides an effective, flexible, and potentially safer strategy to enhance precise genome modifications, which might broadly impact human gene editing and therapy.

15.
ACS Chem Neurosci ; 7(1): 69-81, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26479744

ABSTRACT

A series of novel 1-phenyl-3-hydroxy-4-pyridinone derivatives were designed and synthesized as multifunctional agents for Alzheimer's disease (AD) therapy through incorporation of 3-hydroxy-4-pyridinone moiety from deferiprone into the scaffold of H3 receptor antagonists. Most of these new compounds displayed designed quadruple functions, H3 receptor antagonism, Aß aggregation inhibition, metal ion chelation, and radical scavenging. Especially, the most promising compound 5c displayed nanomolar IC50 values in H3 receptor antagonism with high selectivity, efficient capability to interrupt the formation of Aß(1-42) fibrils, good copper and iron chelating properties, and more potent 2,2'-azino-bis(3-ethyl-benzothiazoline-6-sulfonic acid) radical cation (ABTS(•+)) scavenging activity than Trolox. Further biological evaluation revealed that it did not show obvious cytotoxicity and hERG potassium channel inhibition at micromolar concentration. In addition, compound 5c demonstrated suitable pharmacokinetic properties and acceptable blood-brain barrier (BBB) permeability in vivo. All these results indicate that compound 5c is a potential multifunctional candidate for AD therapy.


Subject(s)
Alzheimer Disease/drug therapy , Antipsychotic Agents/chemistry , Antipsychotic Agents/therapeutic use , Pyridones/chemistry , Pyridones/therapeutic use , Amyloid beta-Peptides/metabolism , Animals , Antipsychotic Agents/chemical synthesis , Antipsychotic Agents/pharmacokinetics , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Chelating Agents/chemistry , Chelating Agents/therapeutic use , Drug Design , Histamine Antagonists/chemical synthesis , Histamine Antagonists/chemistry , Histamine Antagonists/therapeutic use , Humans , Protein Aggregation, Pathological/drug therapy , Pyridones/pharmacokinetics
16.
Mol Neurobiol ; 53(2): 1237-1246, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25613019

ABSTRACT

Valproate exposure is associated with increased risks of autism spectrum disorder. To date, the mechanistic details of disturbance of melatonin receptor subtype 1 (MTNR1A) internalization upon valproate exposure remain elusive. By expressing epitope-tagged receptors (MTNR1A-EGFP) in HEK-293 and Neuro-2a cells, we recorded the dynamic changes of MTNR1A intracellular trafficking after melatonin treatment. Using time-lapse confocal microscopy, we showed in living cells that valproic acid interfered with the internalization kinetics of MTNR1A in the presence of melatonin. This attenuating effect was associated with a decrease in the phosphorylation of PKA (Thr197) and ERK (Thr202/Tyr204). VPA treatment did not alter the whole-cell currents of cells with or without melatonin. Furthermore, fluorescence resonance energy transfer imaging data demonstrated that valproic acid reduced the melatonin-initiated association between YFP-labeled ß-arrestin 2 and CFP-labeled MTNR1A. Together, we suggest that valproic acid influences MTNR1A intracellular trafficking and signaling in a ß-arrestin 2-dependent manner.


Subject(s)
Intracellular Space/metabolism , Receptor, Melatonin, MT1/metabolism , Signal Transduction/drug effects , Valproic Acid/pharmacology , beta-Arrestins/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Endocytosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Melatonin/pharmacology , Mice , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , rab GTP-Binding Proteins/metabolism
17.
Cell Res ; 25(6): 674-90, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25998681

ABSTRACT

Septic encephalopathy (SE) is a critical factor determining sepsis mortality. Vascular inflammation is known to be involved in SE, but the molecular events that lead to the development of encephalopathy remain unclear. Using time-lapse in vivo two-photon laser scanning microscopy, we provide the first direct evidence that cecal ligation and puncture in septic mice induces microglial trafficking to sites adjacent to leukocyte adhesion on inflamed cerebral microvessels. Our data further demonstrate that septic injury increased the chemokine CXCL1 level in brain endothelial cells by activating endothelial P2RX7 and eventually enhanced the binding of Mac-1 (CD11b/CD18)-expressing leukocytes to endothelial ICAM-1. In turn, leukocyte adhesion upregulated endothelial CX3CL1, thereby triggering microglia trafficking to the injured site. The sepsis-induced increase in endothelial CX3CL1 was abolished in CD18 hypomorphic mutant mice. Inhibition of the P2RX7 pathway not only decreased endothelial ICAM-1 expression and leukocyte adhesion but also prevented microglia overactivation, reduced brain injury, and consequently doubled the early survival of septic mice. These results demonstrate the role of the P2RX7 pathway in linking neurovascular inflammation to brain damage in vivo and provide a rationale for targeting endothelial P2RX7 for neurovascular protection during SE.


Subject(s)
Brain Injuries/metabolism , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/metabolism , Macrophage-1 Antigen/metabolism , Receptors, Purinergic P2X7/metabolism , Sepsis-Associated Encephalopathy/metabolism , Animals , Brain Injuries/pathology , Cell Adhesion , Endothelial Cells/pathology , Leukocytes/pathology , Mice , Mice, Mutant Strains , Sepsis-Associated Encephalopathy/pathology
18.
Antioxid Redox Signal ; 21(1): 1-16, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24295341

ABSTRACT

AIMS: Although there is accumulating evidence that increased formation of reactive nitrogen species in cerebral vasculature contributes to the progression of ischemic damage, but the underlying molecular mechanisms remain elusive. Peroxiredoxin 1 (Prx1) can initiate the antioxidant response by scavenging free radicals. Therefore, we tested the hypothesis that Prx1 regulates the susceptibility to nitrosative stress damage during cerebral ischemia in vitro and in vivo. RESULTS: Proteomic analysis in endothelial cells revealed that Prx1 was upregulated after stress-related oxygen-glucose deprivation (OGD). Although peroxynitrite upregulated Prx1 rapidly, this was followed by its polyubiquitination within 6 h after OGD mediated by the E3 ubiquitin ligase E6-associated protein (E6AP). OGD colocalized E6AP with nitrotyrosine in endothelial cells. To assess translational relevance in vivo, mice were studied after middle cerebral artery occlusion (MCAO). This was accompanied by Prx1 ubiquitination and degradation by the activation of E6AP. Furthermore, brain delivery of a lentiviral vector encoding Prx1 in mice inhibited blood-brain barrier leakage and neuronal damage significantly following MCAO. INNOVATION AND CONCLUSIONS: Nitrosative stress during ischemic insult activates E6AP E3 ubiquitin ligase that ubiquitinates Prx1 and subsequently worsens cerebral damage. Thus, targeting the Prx1 antioxidant defense pathway may represent a novel treatment strategy for neurovascular protection in stroke.


Subject(s)
Endothelial Cells/metabolism , Peroxiredoxins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Blood-Brain Barrier/metabolism , Immunohistochemistry , Infarction, Middle Cerebral Artery/metabolism , Male , Mice , Peroxiredoxins/genetics , Proteomics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/physiology
19.
Biomaterials ; 35(1): 530-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24120040

ABSTRACT

The translation of experimental stroke research from the laboratory to successful clinical practice remains a formidable challenge. We previously reported that PEGylated-lipid nanoparticles (PLNs) effectively transport across the blood-brain barrier along with less inflammatory responses. In the present study, PLNs conjugated to Fas ligand antibody that selectively present on brain ischaemic region were used for therapeutic targeting. Fluorescent analysis of the mice brain show that encapsulated 3-n-Butylphthalide (dl-NBP) in PLNs conjugated with Fas ligand antibody effectively delivered to ipsilateral region of ischaemic brain. Furthermore, the confocal immunohistochemical study demonstrated that brain-targeted nanocontainers specifically accumulated on OX42 positive microglia cells in ischaemic region of mice model. Finally, dl-NBP encapsulated nano-drug delivery system is resulted in significant improvements in brain injury and in neurological deficit after ischaemia, with the significantly reduced dosages versus regular dl-NBP. Overall, these data suggests that PLNs conjugated to an antibody specific to the Fas ligand constituted an ideal brain targeting drug delivery system for brain ischaemia.


Subject(s)
Antibodies/administration & dosage , Brain Ischemia/therapy , Fas Ligand Protein/immunology , Lipids/chemistry , Nanoparticles , Polyethylene Glycols/chemistry , Animals , Antibodies/chemistry , Disease Models, Animal , Mice
20.
CNS Neurosci Ther ; 20(9): 816-22, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24712523

ABSTRACT

AIMS: Diabetes mellitus increases the risk of stroke, but the mechanisms are unclear. The present study tested the hypothesis that diabetes mellitus disturbs the brain microcirculation and increases the susceptibility to cerebral damage in a middle cerebral artery occlusion (MCAO) model of ischemia. METHODS: Diabetes was induced by streptozocin in mice expressing green fluorescent protein in endothelial cells (Tie2-GFP mice). Four weeks later, they were subjected to transient (20 min) MCAO. In vivo blood flow was measured by two-photon laser-scanning microscopy (TPLSM) in cerebral arteries, veins, and capillaries. RESULTS: There was a significant decrease in red blood cell (RBC) velocity in capillaries in diabetic mice as assessed by TPLSM, yet the regional cerebral blood flow, as assessed by laser Doppler flowmetry, was maintained. Brain capillary flow developed turbulence after MCAO only in diabetic mice. These mice sustained increased neurological deficits after MCAO which were accompanied by an exaggerated degradation of tight junction proteins and blunted CaMKII phosphorylation in cerebral tissues indicating disruption of the blood-brain barrier and disturbed cognitive potential. CONCLUSION: Diabetic mice are more susceptible to disturbances of cerebral capillary blood flow which may predispose them to neurovascular defects following ischemia.


Subject(s)
Cerebrovascular Circulation/physiology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Analysis of Variance , Animals , Blood Flow Velocity/drug effects , Blood Flow Velocity/physiology , Blood-Brain Barrier/physiopathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Disease Models, Animal , Disease Susceptibility , Erythrocytes/physiology , Green Fluorescent Proteins/genetics , Laser-Doppler Flowmetry , Mice , Mice, Transgenic , Receptor, TIE-2/genetics , Receptors, AMPA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL