Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Colloid Interface Sci ; 642: 193-203, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37004254

ABSTRACT

Compared with other anode materials, Li metal anode has higher capacity density and lower electrode potential, which has been considered as one of the most promising anode materials. However, the unstable solid electrolyte interface (SEI) leads to Li dendrite growth and the infinite volumetric expansion of Li metal, which seriously hinders the stability and cycle life of Li metal batteries (LMBs). Here, a polyurethane elastomer (TPU) material with high elasticity and air stability is used as the artificial SEI of Li metal anode. Its designed synergistic effect of soft chain forging and hard chain segments not only gives TPU artificial SEI layer good electronic insulation, Li ion conductivity, Li dendrite growth inhibition, high elastic modulus and flexibility to adapt to Li volume expansion, but also has a significant air protection effect on the Li metal surface, so that the TPU coated Li foil will not occur obvious oxidation phenomenon after being placed in air for 45 min. The Li symmetric battery modified by TPU achieved a stable and long-term cycle performance of 1300 h at 1 mA/cm2, it can also cycle stably at a high current density of 10 mA/cm2. The Coulomb efficiency of the modified Li/Cu half-cell maintains at above 97% after 400 cycles. In addition, the full cell with LiFePO4 cathode also delivers a very excellent long cycle stability with 90% capacity retention after 1500 cycles at 5 C. This surface modification strategy of SEI on lithium anode has has great research value and will help to improve the widely application of LMBs.

2.
FASEB J ; 36(11): e22619, 2022 11.
Article in English | MEDLINE | ID: mdl-36269280

ABSTRACT

Blood-retinal barrier (BRB) breakdown is responsible for multiple ocular diseases, such as diabetic retinopathy, age-related macular degeneration, and retinal vascular occlusive diseases. Increased vascular permeability contributes to vasogenic edema and tissue damage, with consequent adverse effects on vision. Herein, we found that endothelial CYP2J2 overexpression maintained BRB integrity after ischemia-reperfusion injury and consequently protected against retinal ganglion cell loss. Oxidative stress repressed endothelial ANXA1 expression in vivo and in vitro. CYP2J2 upregulated methyltransferase-like 3 (METTL3) expression and hence promoted ANXA1 translation via ANXA1 m6 A modification in endothelium under oxidative stress. CYP2J2 maintained the distribution of endothelial tight junctions and adherens junctions in an ANXA1-dependent manner. Endothelial ANXA1 plays an indispensable role in vascular homeostasis and stabilization during development. Endothelial ANXA1 deletion disrupted retinal vascular perfusion as well as BRB integrity. CYP2J2 metabolites restored BRB integrity in the presence of ANXA1. Our findings identified the CYP2J2-METTL3-ANXA1 pathway as a potential therapeutic target for relieving BRB impairments.


Subject(s)
Blood-Retinal Barrier , Cytochrome P-450 CYP2J2 , Retinal Diseases , Humans , Annexin A1/genetics , Annexin A1/metabolism , Blood-Retinal Barrier/metabolism , Capillary Permeability , Cytochrome P-450 CYP2J2/genetics , Cytochrome P-450 CYP2J2/metabolism , Diabetic Retinopathy/metabolism , Endothelium/metabolism , Methyltransferases/metabolism , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Ganglion Cells/metabolism , Up-Regulation , Animals , Rats
3.
Food Funct ; 13(22): 11627-11637, 2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36269305

ABSTRACT

Sialyllactose (SL), one of the most abundant oligosaccharides present in porcine breast milk, has been implicated in many biological functions, including the prebiotic and immune-modulating effects. This study was conducted to investigate the influences of dietary SL supplementation on intestinal barrier functions exposure to enterotoxigenic Escherichia coli (ETEC) in a porcine model. Thirty-two pigs were assigned to four treatments, fed with basal or SL-containing (5.0 g kg-1) diet, and orally infused with ETEC or culture medium. SL supplementation significantly reduced the diarrhea incidence and the abundance of E. coli in feces (P < 0.05). Interestingly, SL attenuated ETEC-induced intestinal epithelium injury as indicated by the decreased serum concentrations of diamine oxidase (DAO) and D-lactate and reduced the number of apoptotic cells in the jejunal epithelium (P < 0.05). Moreover, SL not only elevated the abundance of the tight-junction protein ZO-1 in the duodenal and ileal epithelium but also elevated the antioxidant capacity and the number of SIgA positive cells in the jejunal epithelium upon the ETEC challenge (P < 0.05). Importantly, SL decreased the expression levels of inflammation-related genes such as the tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), myeloid differentiation factor 88 (MyD88) in the duodenum, and ileum upon ETEC challenge (P < 0.05). SL also significantly elevated the expression levels of two critical antioxidant genes such as the nuclear factor erythroid-2 related factor 2 (Nrf-2) and kelch-like ECH-associated protein 1 (KEAP-1) in the jejunum (P < 0.05). These results suggested that SL can alleviate ETEC-induced intestinal epithelium injury, which is associated with suppressed inflammation, improved intestinal immunity, antioxidant capacity, and improved intestinal epithelial functions.


Subject(s)
Enterotoxigenic Escherichia coli , Escherichia coli Infections , Swine , Animals , Antioxidants/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/veterinary , Intestinal Mucosa/metabolism , Inflammation/metabolism
4.
Nat Commun ; 13(1): 4345, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35896549

ABSTRACT

Heart failure with reduced ejection fraction (HFrEF) is associated with high mortality, highlighting an urgent need for new therapeutic strategies. As stress-activated cardiac signaling cascades converge on the nucleus to drive maladaptive gene programs, interdicting pathological transcription is a conceptually attractive approach for HFrEF therapy. Here, we demonstrate that CDK7/12/13 are critical regulators of transcription activation in the heart that can be pharmacologically inhibited to improve HFrEF. CDK7/12/13 inhibition using the first-in-class inhibitor THZ1 or RNAi blocks stress-induced transcription and pathologic hypertrophy in cultured rodent cardiomyocytes. THZ1 potently attenuates adverse cardiac remodeling and HFrEF pathogenesis in mice and blocks cardinal features of disease in human iPSC-derived cardiomyocytes. THZ1 suppresses Pol II enrichment at stress-transactivated cardiac genes and inhibits a specific pathologic gene program in the failing mouse heart. These data identify CDK7/12/13 as druggable regulators of cardiac gene transactivation during disease-related stress, suggesting that HFrEF features a critical dependency on transcription that can be therapeutically exploited.


Subject(s)
Cyclin-Dependent Kinases , Heart Failure , Animals , Cell Line, Tumor , Cyclin-Dependent Kinases/genetics , Heart Failure/drug therapy , Heart Failure/genetics , Humans , Mice , RNA Polymerase II , Stroke Volume
5.
Sci Adv ; 8(10): eabj2917, 2022 Mar 11.
Article in English | MEDLINE | ID: mdl-35263131

ABSTRACT

Circulating corticosteroids orchestrate stress adaptation, including inhibition of inflammation. While pathways governing corticosteroid biosynthesis and intracellular signaling are well understood, less is known about mechanisms controlling plasma corticosteroid transport. Here, we show that hepatocyte KLF15 (Kruppel-like factor 15) controls plasma corticosteroid transport and inflammatory responses through direct transcriptional activation of Serpina6, which encodes corticosteroid-binding globulin (CBG). Klf15-deficient mice have profoundly low CBG, reduced plasma corticosteroid binding capacity, and heightened mortality during inflammatory stress. These defects are completely rescued by reconstituting CBG, supporting that KLF15 works primarily through CBG to control plasma corticosterone homeostasis. To understand transcriptional mechanisms, we generated the first KLF15 cistromes using newly engineered Klf153xFLAG mice. Unexpectedly, liver KLF15 is predominantly promoter enriched, including Serpina6, where it binds a palindromic GC-rich motif, opens chromatin, and transactivates genes with minimal associated direct gene repression. Overall, we provide critical mechanistic insight into KLF15 function and identify a hepatocyte-intrinsic transcriptional module that potently regulates systemic corticosteroid transport and inflammation.

6.
J Anim Sci Biotechnol ; 13(1): 30, 2022 Mar 03.
Article in English | MEDLINE | ID: mdl-35236420

ABSTRACT

BACKGROUND: Sialyllactose (SL) is one of the most abundant oligosaccharides present in porcine breast milk. However, little is known about its effect on growth performance and intestinal health in weaned pigs. This study was conducted to explore the protective effect of SL on intestinal epithelium in weaned pigs upon enterotoxigenic Escherichia coli (ETEC) challenge. METHODS: Thirty-two pigs were randomly divided into four treatments. Pigs fed with a basal diet or basal diet containing SL (5.0 g/kg) were orally infused with ETEC or culture medium. RESULTS: SL supplementation elevated the average daily gain (ADG) and feed efficiency in the ETEC-challenged pigs (P < 0.05). SL also improved the digestibilities of dry matter (DM), gross energy (GE), and ash in non-challenged pigs (P < 0.05). Moreover, SL not only elevated serum concentrations of immunoglobulins (IgA, IgG, and IgM), but also significantly decreased the serum concentrations of inflammatory cytokines (TNF-α, IL-1ß, and IL-6) upon ETEC challenge (P < 0.05). Interestingly, SL increased the villus height, the ratio of villus height to crypt depth (V:C), and the activities of mucosal sucrase and maltase in the jejunum and ileum (P < 0.05). SL also elevated the concentrations of microbial metabolites (e.g. acetic acid, propanoic acid, and butyric acid) and the abundance of Lactobacillus, Bifidobacterium, and Bacillus in the cecum (P < 0.05). Importantly, SL significantly elevated the expression levels of jejunal zonula occludins-1 (ZO-1), occluding, and fatty acid transport protein-4 (FATP4) in the ETEC-challenged pigs (P < 0.05). CONCLUSIONS: SL can alleviate inflammation and intestinal injury in weaned pigs upon ETEC challenge, which was associated with suppressed secretion of inflammatory cytokines and elevated serum immunoglobulins, as well as improved intestinal epithelium functions and microbiota.

7.
Cell Death Dis ; 12(12): 1149, 2021 12 11.
Article in English | MEDLINE | ID: mdl-34897284

ABSTRACT

Glaucoma is a leading cause of irreversible blindness worldwide and is characterized by progressive optic nerve degeneration and retinal ganglion cell loss. Axonal transport deficits have been demonstrated to be the earliest crucial pathophysiological changes underlying axonal degeneration in glaucoma. Here, we explored the role of the tetraspanin superfamily member CD82 in an acute ocular hypertension model. We found a transient downregulation of CD82 after acute IOP elevation, with parallel emergence of axonal transport deficits. The overexpression of CD82 with an AAV2/9 vector in the mouse retina improved optic nerve axonal transport and ameliorated subsequent axon degeneration. Moreover, the CD82 overexpression stimulated optic nerve regeneration and restored vision in a mouse optic nerve crush model. CD82 exerted a protective effect through the upregulation of TRAF2, which is an E3 ubiquitin ligase, and activated mTORC1 through K63-linked ubiquitylation and intracellular repositioning of Raptor. Therefore, our study offers deeper insight into the tetraspanin superfamily and demonstrates a potential neuroprotective strategy in glaucoma treatment.


Subject(s)
Axonal Transport , Glaucoma , Animals , Axons/metabolism , Disease Models, Animal , Glaucoma/metabolism , Intraocular Pressure , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Retinal Ganglion Cells
8.
Oxid Med Cell Longev ; 2021: 9397960, 2021.
Article in English | MEDLINE | ID: mdl-34552687

ABSTRACT

OBJECTIVES: In glaucomatous eyes, the main aqueous humor (AH) outflow pathway is damaged by accumulated oxidative stress arising from the microenvironment, vascular dysregulation, and aging, which results in increased outflow resistance and ocular hypertension. Schlemm's canal (SC) serves as the final filtration barrier of the main AH outflow pathway. The present study is aimed at investigating the possible regulation of vasoactive intestinal peptide (VIP) on the cytoskeleton by stabilizing ZO-1 in SC. METHODS: Model of chronic ocular hypertension (COH) induced by episcleral venous cauterization was treated with topical VIP. The ultrastructure of junctions, ZO-1 levels, and permeability of the SC inner wall to FITC-dextran (70 kDa) were detected in the COH models. The F-actin distribution, F/G-actin ratio, and ZO-1 degradation pathway in human umbilical vein endothelial cells (HUVECs) and HEK 293 cells were investigated. RESULTS: ZO-1 in the outer wall of the SC was less than that in the inner wall. COH elicited junction disruption, ZO-1 reduction, and increased permeability of the SC inner wall to FITC-dextran in rats. ZO-1 plays an essential role in maintaining the F/G-actin ratio and F-actin distribution. VIP treatment attenuated the downregulation of ZO-1 associated with COH or H2O2-induced oxidative damage. In H2O2-stimulated HUVECs, the caspase-3 inhibitor prevents ZO-1 disruption. Caspase-3 activation promoted endolysosomal degradation of ZO-1. Furthermore, a decrease in caspase-3 activation and cytoskeleton redistribution was demonstrated in VIP + H2O2-treated cells. The knockdown of ZO-1 or the overexpression of caspase-3 blocked the effect of VIP on the cytoskeleton. CONCLUSION: This study provides insights into the role of VIP in stabilizing the interaction between the actin cytoskeleton and cell junctions and may provide a promising targeted strategy for glaucoma treatment.


Subject(s)
Actin Cytoskeleton/chemistry , Caspase 3/metabolism , Endothelium, Vascular/metabolism , Glaucoma/metabolism , Sclera/metabolism , Vasoactive Intestinal Peptide/pharmacology , Zonula Occludens-1 Protein/metabolism , Animals , Aqueous Humor/drug effects , Aqueous Humor/metabolism , Caspase 3/genetics , Endosomes/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Glaucoma/drug therapy , Glaucoma/pathology , Lysosomes/metabolism , Male , Rats , Rats, Sprague-Dawley , Sclera/drug effects , Sclera/pathology , Zonula Occludens-1 Protein/genetics
9.
Nature ; 595(7867): 438-443, 2021 07.
Article in English | MEDLINE | ID: mdl-34163071

ABSTRACT

In diseased organs, stress-activated signalling cascades alter chromatin, thereby triggering maladaptive cell state transitions. Fibroblast activation is a common stress response in tissues that worsens lung, liver, kidney and heart disease, yet its mechanistic basis remains unclear1,2. Pharmacological inhibition of bromodomain and extra-terminal domain (BET) proteins alleviates cardiac dysfunction3-7, providing a tool to interrogate and modulate cardiac cell states as a potential therapeutic approach. Here we use single-cell epigenomic analyses of hearts dynamically exposed to BET inhibitors to reveal a reversible transcriptional switch that underlies the activation of fibroblasts. Resident cardiac fibroblasts demonstrated robust toggling between the quiescent and activated state in a manner directly correlating with BET inhibitor exposure and cardiac function. Single-cell chromatin accessibility revealed previously undescribed DNA elements, the accessibility of which dynamically correlated with cardiac performance. Among the most dynamic elements was an enhancer that regulated the transcription factor MEOX1, which was specifically expressed in activated fibroblasts, occupied putative regulatory elements of a broad fibrotic gene program and was required for TGFß-induced fibroblast activation. Selective CRISPR inhibition of the single most dynamic cis-element within the enhancer blocked TGFß-induced Meox1 activation. We identify MEOX1 as a central regulator of fibroblast activation associated with cardiac dysfunction and demonstrate its upregulation after activation of human lung, liver and kidney fibroblasts. The plasticity and specificity of BET-dependent regulation of MEOX1 in tissue fibroblasts provide previously unknown trans- and cis-targets for treating fibrotic disease.


Subject(s)
Enhancer Elements, Genetic , Fibroblasts/cytology , Heart Diseases/genetics , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Chromatin/metabolism , Epigenomics , Gene Expression Regulation , Humans , Mice , Proteins/antagonists & inhibitors , Single-Cell Analysis , Transcriptome , Transforming Growth Factor beta/metabolism
10.
Int J Mol Sci ; 22(7)2021 Mar 27.
Article in English | MEDLINE | ID: mdl-33801629

ABSTRACT

The Na/K-ATPase is the specific receptor for cardiotonic steroids (CTS) such as ouabain and digoxin. At pharmacological concentrations used in the treatment of cardiac conditions, CTS inhibit the ion-pumping function of Na/K-ATPase. At much lower concentrations, in the range of those reported for endogenous CTS in the blood, they stimulate hypertrophic growth of cultured cardiac myocytes through initiation of a Na/K-ATPase-mediated and reactive oxygen species (ROS)-dependent signaling. To examine a possible effect of endogenous concentrations of CTS on cardiac structure and function in vivo, we compared mice expressing the naturally resistant Na/K-ATPase α1 and age-matched mice genetically engineered to express a mutated Na/K-ATPase α1 with high affinity for CTS. In this model, total cardiac Na/K-ATPase activity, α1, α2, and ß1 protein content remained unchanged, and the cardiac Na/K-ATPase dose-response curve to ouabain shifted to the left as expected. In males aged 3-6 months, increased α1 sensitivity to CTS resulted in a significant increase in cardiac carbonylated protein content, suggesting that ROS production was elevated. A moderate but significant increase of about 15% of the heart-weight-to-tibia-length ratio accompanied by an increase in the myocyte cross-sectional area was detected. Echocardiographic analyses did not reveal any change in cardiac function, and there was no fibrosis or re-expression of the fetal gene program. RNA sequencing analysis indicated that pathways related to energy metabolism were upregulated, while those related to extracellular matrix organization were downregulated. Consistent with a functional role of the latter, an angiotensin-II challenge that triggered fibrosis in the α1r/rα2s/s mouse failed to do so in the α1s/sα2s/s. Taken together, these results are indicative of a link between circulating CTS, Na/K-ATPase α1, ROS, and physiological cardiac hypertrophy in mice under baseline laboratory conditions.


Subject(s)
Cardiac Glycosides/chemistry , Heart/physiology , Myocardium/enzymology , Sodium-Potassium-Exchanging ATPase/genetics , Angiotensin II/pharmacology , Animals , Cardiomegaly/pathology , Disease Models, Animal , Echocardiography , Heart/drug effects , Male , Mice , Mutation , Ouabain/pharmacology , Protein Isoforms , RNA-Seq , Reactive Oxygen Species , Signal Transduction/drug effects
11.
Circulation ; 142(24): 2338-2355, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33094644

ABSTRACT

BACKGROUND: Gene regulatory networks control tissue homeostasis and disease progression in a cell type-specific manner. Ubiquitously expressed chromatin regulators modulate these networks, yet the mechanisms governing how tissue specificity of their function is achieved are poorly understood. BRD4 (bromodomain-containing protein 4), a member of the BET (bromo- and extraterminal domain) family of ubiquitously expressed acetyl-lysine reader proteins, plays a pivotal role as a coactivator of enhancer signaling across diverse tissue types in both health and disease and has been implicated as a pharmacological target in heart failure. However, the cell-specific role of BRD4 in adult cardiomyocytes remains unknown. METHODS: We combined conditional mouse genetics, unbiased transcriptomic and epigenomic analyses, and classic molecular biology and biochemical approaches to understand the mechanism by which BRD4 in adult cardiomyocyte homeostasis. RESULTS: Here, we show that cardiomyocyte-specific deletion of Brd4 in adult mice leads to acute deterioration of cardiac contractile function with mutant animals demonstrating a transcriptomic signature characterized by decreased expression of genes critical for mitochondrial energy production. Genome-wide occupancy data show that BRD4 enriches at many downregulated genes (including the master coactivators Ppargc1a, Ppargc1b, and their downstream targets) and preferentially colocalizes with GATA4 (GATA binding protein 4), a lineage-determining cardiac transcription factor not previously implicated in regulation of adult cardiac metabolism. BRD4 and GATA4 form an endogenous complex in cardiomyocytes and interact in a bromodomain-independent manner, revealing a new functional interaction partner for BRD4 that can direct its locus and tissue specificity. CONCLUSIONS: These results highlight a novel role for a BRD4-GATA4 module in cooperative regulation of a cardiomyocyte-specific gene program governing bioenergetic homeostasis in the adult heart.


Subject(s)
Energy Metabolism , GATA4 Transcription Factor/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Ventricular Dysfunction, Left/metabolism , Animals , Energy Metabolism/genetics , GATA4 Transcription Factor/genetics , Gene Expression Profiling , Gene Expression Regulation , Genotype , HEK293 Cells , Homeostasis , Humans , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/genetics , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/ultrastructure , Nuclear Proteins/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phenotype , Protein Binding , Rats, Sprague-Dawley , Transcription Factors/genetics , Transcriptome , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left
12.
J Clin Invest ; 130(6): 2966-2977, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32106109

ABSTRACT

Salt-inducible kinases (SIKs) are key regulators of cellular metabolism and growth, but their role in cardiomyocyte plasticity and heart failure pathogenesis remains unknown. Here, we showed that loss of SIK1 kinase activity protected against adverse cardiac remodeling and heart failure pathogenesis in rodent models and cardiomyocytes derived from human induced pluripotent stem cells. We found that SIK1 phosphorylated and stabilized histone deacetylase 7 (HDAC7) protein during cardiac stress, an event that is required for pathologic cardiomyocyte remodeling. Gain- and loss-of-function studies of HDAC7 in cultured cardiomyocytes implicated HDAC7 as a prohypertrophic signaling effector that can induce c-Myc expression, indicating a functional departure from the canonical MEF2 corepressor function of class IIa HDACs. Taken together, our findings reveal what we believe to be a previously unrecognized role for a SIK1/HDAC7 axis in regulating cardiac stress responses and implicate this pathway as a potential target in human heart failure.


Subject(s)
Histone Deacetylases/metabolism , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Ventricular Remodeling , Animals , Humans , Mice , Myocytes, Cardiac/pathology , Phosphorylation , Rats , Rats, Sprague-Dawley
13.
Development ; 146(23)2019 12 09.
Article in English | MEDLINE | ID: mdl-31784461

ABSTRACT

Long intergenic non-coding RNAs (lincRNAs) have been implicated in gene regulation, but their requirement for development needs empirical interrogation. We computationally identified nine murine lincRNAs that have developmentally regulated transcriptional and epigenomic profiles specific to early heart differentiation. Six of the nine lincRNAs had in vivo expression patterns supporting a potential function in heart development, including a transcript downstream of the cardiac transcription factor Hand2, which we named Handlr (Hand2-associated lincRNA), Rubie and Atcayos We genetically ablated these six lincRNAs in mouse, which suggested genomic regulatory roles for four of the cohort. However, none of the lincRNA deletions led to severe cardiac phenotypes. Thus, we stressed the hearts of adult Handlr and Atcayos mutant mice by transverse aortic banding and found that absence of these lincRNAs did not affect cardiac hypertrophy or left ventricular function post-stress. Our results support roles for lincRNA transcripts and/or transcription in the regulation of topologically associated genes. However, the individual importance of developmentally specific lincRNAs is yet to be established. Their status as either gene-like entities or epigenetic components of the nucleus should be further considered.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Heart/embryology , Myocardium/metabolism , RNA, Long Noncoding/biosynthesis , Animals , Gene Deletion , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Mice , Mice, Mutant Strains , RNA, Long Noncoding/genetics
14.
FASEB J ; 33(10): 11194-11209, 2019 10.
Article in English | MEDLINE | ID: mdl-31295013

ABSTRACT

Glaucoma is a leading cause of irreversible blindness worldwide. Vascular factors play a substantial role in the pathogenesis of glaucoma. Expressed in the vascular endothelium, cytochrome P450 (CYP) 2J2 is one of the CYP epoxygenases that metabolize arachidonic acid to produce epoxyeicosatrienoic acids and exert pleiotropic protective effects on the vasculature. In the present study, we investigated whether endothelium-specific overexpression of CYP2J2 (tie2-CYP2J2-Tr) protects against retinal ganglion cell (RGC) loss induced by glaucoma and in what way retinal vessels are involved in this process. We used a glaucoma model of retinal ischemia-reperfusion (I/R) injury in rats and found that endothelium-specific overexpression of CYP2J2 attenuated RGC loss induced by retinal I/R. Moreover, retinal I/R triggered retinal vascular senescence, indicated by up-regulated senescence-related proteins p53, p16, and ß-galactosidase activity. The senescent endothelial cells resulted in pericyte loss and increased endothelial secretion of matrix metallopeptidase 9, which further contributed to RGC loss. CYP2J2 overexpression alleviated vascular senescence, pericyte loss, and matrix metallopeptidase 9 secretion. CYP2J2 suppressed endothelial senescence by down-regulating senescence-associated proteins p53 and p16. These 2 proteins were positively regulated by microRNA-128-3p, which was inhibited by CYP2J2. These results suggest that CYP2J2 protects against endothelial senescence and RGC loss in glaucoma, a discovery that may lead to the development of a potential treatment strategy for glaucoma.-Huang, J., Zhao, Q., Li, M., Duan, Q., Zhao, Y., Zhang, H. The effects of endothelium-specific CYP2J2 overexpression on the attenuation of retinal ganglion cell apoptosis in a glaucoma rat model.


Subject(s)
Apoptosis/physiology , Cytochrome P-450 Enzyme System/metabolism , Endothelium, Vascular/metabolism , Glaucoma/metabolism , Retinal Ganglion Cells/metabolism , Animals , Cellular Senescence/physiology , Cytochrome P-450 CYP2J2 , Disease Models, Animal , Down-Regulation/physiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Glaucoma/pathology , Metalloendopeptidases/metabolism , MicroRNAs/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Retinal Ganglion Cells/pathology , Tumor Suppressor Protein p53/metabolism , Up-Regulation/physiology
15.
Front Cell Neurosci ; 12: 347, 2018.
Article in English | MEDLINE | ID: mdl-30364320

ABSTRACT

Variants near the ATP-binding cassette transporter A1 (ABCA1) gene are associated with elevated intraocular pressure and newly discovered risk factors for glaucoma. Previous studies have shown an association between ABCA1 deficiency and retinal inflammation. Using a mouse model of ischemia-reperfusion (IR) induced by acute intraocular pressure elevation, we found that the retinal expression of ABCA1 protein was decreased. An induction of ABCA1 expression by liver X receptor agonist TO901317 reduced retinal ganglion cell (RGC) apoptosis after IR and promoted membrane translocation and secretion of the anti-inflammatory factor annexin A1 (ANXA1). Moreover, ABCA1 and ANXA1 co-localized in cell membranes, and the interaction domain is amino acid 196 to 274 of ANXA1 fragment. TO901317 also reduced microglia migration and activation and decreased the expression of pro-inflammatory cytokines interleukin (IL)-17A and IL-1ß, which could be reversed by the ANXA1 receptor blocker Boc2. Overexpression of TANK-binding kinase 1 (TBK1) increased ABCA1 degradation, which was reversed by the proteasome inhibitor carbobenzoxy-L-leucyl-L-leucyl-L-leucinal (MG132). Silencing Tbk1 with siRNA increased ABCA1 expression and promoted ANXA1 membrane translocation. These results indicate a novel IR mechanism, that leads via TBK1 activation to ABCA1 ubiquitination. This degradation decreases ANXA1 secretion, thus facilitating retinal inflammation and RGC apoptosis. Our findings suggest a potential treatment strategy to prevent RGC apoptosis in retinal ischemia and glaucoma.

16.
J Cardiovasc Pharmacol ; 71(2): 95-103, 2018 02.
Article in English | MEDLINE | ID: mdl-29419572

ABSTRACT

Ouabain preconditioning (OPC) initiated by low concentrations of the cardiac glycoside (CG) ouabain binding to Na/K-ATPase is relayed by a unique intracellular signaling and protects cardiac myocytes against ischemia/reperfusion injury. To explore more clinically applicable protocols based on CG properties, we tested whether the FDA-approved CG digoxin could trigger cardioprotective effects comparable with those of ouabain using PC, preconditioning and PostC, postconditioning protocols in the Langendorff-perfused mouse heart subjected to global ischemia and reperfusion. Ouabain or digoxin at 10 µmol/L inhibited Na/K-ATPase activity by approximately 30% and activated PKCε translocation by approximately 50%. Digoxin-induced PC (DigPC), initiated by a transient exposure before 40 minutes of ischemia, was as effective as OPC as suggested by the recovery of left ventricular developed pressure, end-diastolic pressure, and cardiac Na/K-ATPase activity after 30 minutes of reperfusion. DigPC also significantly decreased lactate dehydrogenase release and reduced infarct size, comparable with OPC. PostC protocols consisting of a single bolus injection of 100 nmoles of ouabain or digoxin in the coronary tree at the beginning of reperfusion both improved significantly the recovery of left ventricular developed pressure and decreased lactate dehydrogenase release, demonstrating a functional and structural protection comparable with the one provided by OPC. Given the unique signaling triggered by OPC, these results suggest that DigPostC could be considered for patients with risk factors and/or concurrent treatments that may limit effectiveness of ischemic PostC.


Subject(s)
Cardiotonic Agents/administration & dosage , Digoxin/administration & dosage , Enzyme Inhibitors/administration & dosage , Myocardial Contraction/drug effects , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Ouabain/administration & dosage , Ventricular Function, Left/drug effects , Animals , Cell Death/drug effects , Disease Models, Animal , Isolated Heart Preparation , Male , Mice, Inbred C57BL , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Protein Kinase C-epsilon/metabolism , Recovery of Function , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism , Ventricular Pressure/drug effects
17.
Brain Res ; 1675: 78-86, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28893580

ABSTRACT

Primary open angle glaucoma (POAG) is a leading cause of irreversible blindness worldwide. Previous MRI studies have revealed that POAG can be associated with alterations in hippocampal function. Thus, the aim of this study was to investigate a relationship between chronic high intraocular pressure (IOP) and hippocampal changes in a rat model. We used behavioural tests to assess learning and memory ability, and additionally investigated the hippocampal expression of pathological amyloid beta (Aß), phospho-tau, and related pathway proteins. Chronic high IOP impaired learning and memory in rats and concurrently increased Aß and phospho-tau expression in the hippocampus by altering the activation of different kinase (GSK-3ß, BACE1) and phosphatase (PP2A) proteins in the hippocampus. This study provides novel evidence for the relationship between high IOP and hippocampal alterations, especially in the context of learning and memory.


Subject(s)
Intraocular Pressure/physiology , Maze Learning/physiology , Memory Disorders/pathology , Ocular Hypertension/pathology , Animals , Hippocampus/metabolism , Hippocampus/pathology , Male , Memory Disorders/etiology , Memory Disorders/metabolism , Ocular Hypertension/complications , Ocular Hypertension/metabolism , Rats , Rats, Sprague-Dawley
18.
Sci Transl Med ; 9(390)2017 05 17.
Article in English | MEDLINE | ID: mdl-28515341

ABSTRACT

Despite current standard of care, the average 5-year mortality after an initial diagnosis of heart failure (HF) is about 40%, reflecting an urgent need for new therapeutic approaches. Previous studies demonstrated that the epigenetic reader protein bromodomain-containing protein 4 (BRD4), an emerging therapeutic target in cancer, functions as a critical coactivator of pathologic gene transactivation during cardiomyocyte hypertrophy. However, the therapeutic relevance of these findings to human disease remained unknown. We demonstrate that treatment with the BET bromodomain inhibitor JQ1 has therapeutic effects during severe, preestablished HF from prolonged pressure overload, as well as after a massive anterior myocardial infarction in mice. Furthermore, JQ1 potently blocks agonist-induced hypertrophy in human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Integrated transcriptomic analyses across animal models and human iPSC-CMs reveal that BET inhibition preferentially blocks transactivation of a common pathologic gene regulatory program that is robustly enriched for NFκB and TGF-ß signaling networks, typified by innate inflammatory and profibrotic myocardial genes. As predicted by these specific transcriptional mechanisms, we found that JQ1 does not suppress physiological cardiac hypertrophy in a mouse swimming model. These findings establish that pharmacologically targeting innate inflammatory and profibrotic myocardial signaling networks at the level of chromatin is effective in animal models and human cardiomyocytes, providing the critical rationale for further development of BET inhibitors and other epigenomic medicines for HF.


Subject(s)
Cardiomegaly/metabolism , Heart Failure/metabolism , Inflammation/metabolism , Proteins/metabolism , Animals , Azepines/therapeutic use , Cardiomegaly/drug therapy , Cardiomegaly/genetics , Gene Regulatory Networks/genetics , Gene Regulatory Networks/physiology , Heart Failure/drug therapy , Heart Failure/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Inflammation/genetics , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Proteins/genetics , Signal Transduction , Transforming Growth Factor beta/metabolism , Triazoles/therapeutic use
19.
Am J Physiol Cell Physiol ; 312(3): C222-C232, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-27903584

ABSTRACT

The Na/K-ATPase α1 polypeptide supports both ion-pumping and signaling functions. The Na/K-ATPase α3 polypeptide differs from α1 in both its primary structure and its tissue distribution. The expression of α3 seems particularly important in neurons, and recent clinical evidence supports a unique role of this isoform in normal brain function. The nature of this specific role of α3 has remained elusive, because the ubiquitous presence of α1 has hindered efforts to characterize α3-specific functions in mammalian cell systems. Using Na/K-ATPase α1 knockdown pig kidney cells (PY-17), we generated the first stable mammalian cell line expressing a ouabain-resistant form of rat Na/K-ATPase α3 in the absence of endogenous pig α1 detectable by Western blotting. In these cells, Na/K-ATPase α3 formed a functional ion-pumping enzyme and rescued the expression of Na/K-ATPase ß1 and caveolin-1 to levels comparable with those observed in PY-17 cells rescued with a rat Na/K-ATPase α1 (AAC-19). The α3-containing enzymes had lower Na+ affinity and lower ouabain-sensitive transport activity than their α1-containing counterparts under basal conditions, but showed a greater capacity to be activated when intracellular Na+ was increased. In contrast to Na/K-ATPase α1, α3 could not regulate Src. Upon exposure to ouabain, Src activation did not occur, yet ERK was activated through Src-independent pathways involving PI3K and PKC. Hence, α3 expression confers signaling and pumping properties that are clearly distinct from that of cells expressing Na/K-ATPase α1.


Subject(s)
Ion Channel Gating/physiology , Kidney/metabolism , MAP Kinase Signaling System/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/metabolism , Stem Cells/enzymology , src-Family Kinases/metabolism , Animals , Cell Line , Cell Proliferation/physiology , Kidney/cytology , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Rats
20.
Physiol Rep ; 4(19)2016 10.
Article in English | MEDLINE | ID: mdl-27702882

ABSTRACT

Cardiac glycosides (CG) are traditionally known as positive cardiac inotropes that inhibit Na+/K+-ATPase-dependent ion transport. CG also trigger-specific signaling pathways through the cardiac Na+/K+-ATPase, with beneficial effects in ischemia/reperfusion (I/R) injury (e.g., ouabain preconditioning, known as OPC) and hypertrophy. Our current understanding of hypersensitivity to CG and subsequent toxicity in the ischemic heart is mostly based on specific I/R-induced alterations of the Na+/K+-ATPase enzymatic function and has remained incomplete. The primary goal of this study was to investigate and compare the impact of I/R on Na+/K+-ATPase enzymatic and signaling functions. Second, we assessed the impact of OPC on both functions. Langendorff-perfused rat hearts were exposed to 30 min of ischemia and 30 min of reperfusion. At the inotropic concentration of 50 µmol/L, ouabain increased ERK and Akt phosphorylation in control hearts. In I/R hearts, this concentration did not induced positive inotropy and failed to induce Akt or ERK phosphorylation. The inotropic response to dobutamine as well as insulin signaling persisted, suggesting specific alterations of Na+/K+-ATPase. Indeed, Na+/K+-ATPase protein expression was intact, but the enzyme activity was decreased by 60% and the enzymatic function of the isoform with high affinity for ouabain was abolished following I/R. Strikingly, OPC prevented all I/R-induced alterations of the receptor. Further studies are needed to reveal the respective roles of I/R-induced modulations of Na+/K+-ATPase enzymatic and signaling functions in cardiomyocyte death.


Subject(s)
Cardiotonic Agents/pharmacology , Ischemic Preconditioning, Myocardial/methods , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Ouabain/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cardiotonic Agents/administration & dosage , Male , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/drug therapy , Myocardium/enzymology , Myocytes, Cardiac/metabolism , Ouabain/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...